EP1527179B1 - Il-7 als wirkstoff, il-7-haltige zusammensetzung, herstellungsverfahren und verwendungen - Google Patents

Il-7 als wirkstoff, il-7-haltige zusammensetzung, herstellungsverfahren und verwendungen Download PDF

Info

Publication number
EP1527179B1
EP1527179B1 EP03792262A EP03792262A EP1527179B1 EP 1527179 B1 EP1527179 B1 EP 1527179B1 EP 03792262 A EP03792262 A EP 03792262A EP 03792262 A EP03792262 A EP 03792262A EP 1527179 B1 EP1527179 B1 EP 1527179B1
Authority
EP
European Patent Office
Prior art keywords
conformer
composition according
human
anyone
recombinant
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP03792262A
Other languages
English (en)
French (fr)
Other versions
EP1527179A2 (de
Inventor
Michel Christian Morre
Brigitte Assouline
Pierre Cortez
Anne Gregoire
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cytheris SA
Original Assignee
Cytheris SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cytheris SA filed Critical Cytheris SA
Priority to EP03792262A priority Critical patent/EP1527179B1/de
Priority to SI200331907T priority patent/SI1527179T1/sl
Publication of EP1527179A2 publication Critical patent/EP1527179A2/de
Application granted granted Critical
Publication of EP1527179B1 publication Critical patent/EP1527179B1/de
Priority to CY20101101165T priority patent/CY1110994T1/el
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5418IL-7
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/022Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from an adenovirus

Definitions

  • the present invention relates, generally, to the fields of immunology and molecular biology.
  • the invention discloses, more particularly, new and improved interleukin-7 drug substances, corresponding specific immunoreactive antibodies, as well as compositions comprising the same, their preparation and uses.
  • the invention also discloses methods to characterize the impurity profile of a r-hIL-7 drug substance used for therapeutic purpose, as well as optimized nucleotide sequences encoding mammalian IL-7, recombinant expression vectors and methods for preparing and purifying said polypeptides.
  • B and T lymphocytes are the primary effector cells of the immune responses. Both cell classes are considered to derive ultimately from hematopoietic stem cells in the mammalian bone marrow, via progenitor or precursor cells representing distinguishable stages in the differentiation of each class. Mature T cells develop principally in the thymus, presumably from a precursor cell which migrates from the bone marrow to the thymus at an early stage of T lymphocyte development. Numbers of factors are active on mature peripheral B and T cells, including IL-1, IL-2, IL-4, IL-5, interferon gamma, BSF-2, neuroleukin, transforming growth factor beta and IL-7 ( EP0314415 ).
  • Interleukin-7 or "IL-7° refers to a mammalian endogenous secretory glycoprotein which is capable of inducing proliferation of bone marrow-derived lymphocyte progenitors and precursors, including the specialized precursors known as pre-B cells. Originally derived from the stromal element of a bone marrow cell line, IL-7 is also secreted by thymic cells, intestinal epithelial cells, keratinocytes and generally all lymphoid tissues. EP0314415 describes mammalian interleukin-7 proteins and DNAs encoding said proteins.
  • IL-7 The mature human IL-7 (r-hIL-7), exclusive of glycosylation, produced in Escherichia coli, which is described in EP0314415 , exhibits a 17,387 Daltons molecular weight and displays a high activity in vitro on specific bioassays based on the proliferation of various lymphocytes populations.
  • Alternative designations for this molecule are "pre-B cell growth factor” and "lymphopoietin-1".
  • IL-7 was disclosed as a cytokine whose principal activity was induction of precursor B cell proliferation ( Namen A.E. et al.; Journal of Experimental medicine; 1988; 167:988-1002 ).
  • IL-7 has more recently been disclosed as being involved in the survival and proliferation of thymocytes (T-Cells) during early stage of T-cell development ( Schluns K.S. et al.; Nature Immunology; 2000; 1(5):426-432 ). Fry and collaborators further identified IL-7 as a potent modulator of thymic-independent T-cell regeneration in a multifactorial action ( Fry T.J. et al.; Blood; 2001; 97(6):1525-1533 ).
  • IL-7 has thus a great potential therapeutic use in the stimulation of the proliferation of T cell precursors, of antibody-secreting B cells, in the stimulation of antigen driven T-Cell peripheral expansion, and in the production of na ⁇ ve T-Cells as well as other hematopoietic cell types.
  • a particularly interesting therapeutic use of active IL-7 molecules is for immune reconstitution of lymphopenic patients: patients treated for a cancer, patients having received a bone marrow or a Stem Cell transfer, patients presenting an acquired or genetic immune deficiency, elderly patients or any patients having low CD4 count.
  • Other interests reside in the ability of IL-7 to produce new na ⁇ ve CD4 T-Cells or to expand specific pools in order to produce or increase specific immune responses (Vaccine enhancement).
  • a drug substance or pharmaceutical composition aimed at stimulating a global or specific immune reconstitution should be efficient on a long term, which implies that it should not trigger a specific immune reaction against its own active principle.
  • the present invention now shows, unexpectedly, that the long term activity of recombinant human IL-7 is mostly expressed by a specific 1-4; 2-5; 3-6 conformer.
  • the present invention further shows that efficient drug substances should not only contain the above conformer as the major constituent, but should also be essentially devoid of other conformers or IL-7 molecular variants, previously considered as active products.
  • the present invention thus discloses improved methods for the preparation and uses of said purified conformer of human IL-7, devoid of the above mentioned product related substances or impurities, which exhibits a particularly advantageous long term in vivo activity, able to favor global or specific immune responses.
  • IL-7 conformer comprising the following three disulfide bridges: Cys: 1-4 (Cys2- Cys92); 2-5 (Cys34- Cys129); 3-6 (Cys47- Cys141).
  • the IL-7 conformer is preferably a human recombinant IL-7 conformer, and may be glycosylated or unglycosylated.
  • a drug substance comprising an IL-7 conformer as described above and essentially devoid of product-related impurities and product related substances, particularly of other conformers or dimers of IL-7.
  • drug product a medicament
  • pharmaceutical composition a drug substance as described above in the manufacture of a medicament (drug product) or pharmaceutical composition.
  • composition comprising an effective amount of a drug substance as described above and one or more pharmaceutically compatible carriers or excipients.
  • nucleic acid molecules coding for an IL-7 polypeptide comprise a sequence encoding an IL-7 polypeptide, said sequence being optimized for the expression, in a recombinant host, of biologically active IL-7, particularly of an IL-7 conformer according to the invention. More specifically, the nucleic acid molecules provide for a limited expression of truncated IL-7 polypeptides and increase the yields of production of biologically active human IL-7 conformer. Particular sequences comprise a mutated or inactivated Shine-Dalgarno like sequence (e.g., SEQ ID NO: 1).
  • sequences comprise a peptide signal causing efficient secretion of the polypeptide (e.g., SEQ ID NO: 3, 16, 18, 20 or 22).
  • the invention also discloses, for the first time, the nucleic acid sequence of the simian IL-7 (e.g., SEQ ID NO: 12, 20 or 22).
  • the invention further discloses vectors comprising a nucleic acid as mentioned above and recombinant host cells comprising said nucleic acid or said vector.
  • the nucleic acids and vectors may be used to produce recombinant mammalian IL-7 polypeptides in various competent host cells, as well as for gene therapy purposes.
  • the invention also discloses an antibody specifically immunoreactive with a human IL-7 conformer according to the invention, hybridoma cell lines that produce said monoclonal antibody, as well as compositions suitable for diagnosis, assay or therapy comprising said antibody.
  • Disclosed is also a method of producing a (recombinant) IL-7 conformer as described above, from prokaryotic or eukaryotic host cells, as well as a method of detecting or measuring the presence of an IL-7 conformer in a sample or to characterize a sample.
  • the method of producing an IL-7 conformer or a drug substance as defined above comprises:
  • the invention also discloses a method of characterizing or qualifying a sample, comprising detecting or measuring, in said sample, the presence of an IL-7 conformer as described above. Such method is particularly suited and important for validation of clinical lots or batches, whereby samples comprising excessive IL-7 molecular variants and/or product related impurities are discarded or further treated.
  • the invention also discloses to the use of an IL-7 conformer obtained by a method as described above, for the manufacture of a pharmaceutical composition to cause or modulate an immune response in a subject, particularly to induce a prolonged lymphopoiesis stimulation and/or to amplify an immune response.
  • the invention also discloses the use of an IL-7 conformer obtained by a method as described above, for the manufacture of a pharmaceutical composition to prevent or treat a disease associated with an immunodeficiency.
  • the invention further discloses the use of an IL-7 conformer according to the invention as a tool for experimental and pharmacological use in monkeys.
  • the present invention discloses a purified IL-7 conformer, said IL-7 conformer comprising the following three disulfide bridges: Cys: 1-4 (Cys2- Cys92); 2-5 (Cys34- Cys129); 3-6 (Cys47- Cys141) drug substances and pharmaceutical compositions comprising said IL-7 conformer as well as methods for their production and therapeutic uses.
  • the IL-7 conformer is preferably a human recombinant IL-7 conformer, and may be glycosylated or unglycosylated.
  • the human recombinant IL-7 conformer is preferentially glycosylated.
  • the invention is based on the unexpected discovery that the mid or long term in vivo biological activity of IL-7 is essentially attributable to a particular conformation of the molecule, not suspected so far, and that highly improved therapeutic efficiency is achieved by producing pharmaceutical compositions comprising said IL-7 conformer in a substantially pure form.
  • IL-7 conformer comprising the following three disulfide bridges: Cys: 1-4 (Cys2-Cys92); 2-5 (Cys34- Cys129); 3-6 (Cys47- Cys141).
  • IL-7 primary sequence human and simian contains six highly conserved cysteine residues which are involved in the formation of three intramolecular disulfide bridges. As originally described by Goodwin, these disulfide bridges are necessary for bioactivity, beta-mercaptoethanol treatment of the IL-7 molecule fully abolishing its activity ( Goodwin R.G. et al.; Proceedings of the National Academy of Sciences of the USA; 1989; 86:302-306 ). From this point, according to the rank of the cysteine residues positions, various conformers are possible, among which : 1-6; 2-5; 3-4 or 1-4; 2-5; 3-6 or 1-4; 2-6; 3-5 or 1-5; 2-4; 3-6, plus various other conformers, including contiguous bridges.
  • the present invention now shows that the above three-dimensional structure, presented, demonstrated and accepted in the data bank until now, is incorrect and that the long term activity of recombinant human IL-7 is mostly expressed by a specific 1-4; 2-5; 3-6 conformer.
  • the experimental results obtained differ from the previously reported disulfide bonds in human IL-7 (hIL-7) ( Cosenza L. et al.; Journal of Biological Chemistry; 1997; 272:32995-33000 ) and show, for the first time, that long term active IL-7 compositions require the presence of a substantially pure particular IL-7 conformer which is in accordance with other models designed by homology modeling with GM-CSF and IL-4 conformations.
  • an "IL-7 polypeptide” designates a mammalian (e.g., human or simian) IL-7 polypeptide and, more preferably, a human IL-7 polypeptide, especially for uses as therapeutic or vaccine, or a simian IL-7 polypeptide, especially for use in non human primate experiments.
  • Preferred IL-7 polypeptides of this invention comprise an amino acid sequence as described in EP 314 415 as well as any natural variants and homologs thereof.
  • the sequence of human IL-7 is available on genebanks.
  • the typical wild-type protein comprises 152 amino acids and, optionally, an additional N-terminal methionine residue (SEQ ID Nos: 2, 4, 17 and 19).
  • Variants thereof include, more preferably, natural allelic variants resulting from natural polymorphism, including SNPs, splicing variants, etc.
  • the term IL-7 polypeptide is meant to designate a polypeptide having the sequence of SEQ ID NO: 2, 13 [SEQ ID NO: 13 comprising or not the signal peptide (amino acids no: 2-25)], 17, 19, 21 or 23, as well as natural variants thereof.
  • the specific IL-7 polypeptide used in the present invention is preferably a recombinant IL-7, more preferably a recombinant human IL-7 conformer comprising the amino acid sequence SEQ ID NO: 2, 4, 17 or 19.
  • a specific "conformer" of IL-7 designates an IL-7 polypeptide having a particular three-dimensional configuration.
  • recombinant means that the polypeptide is obtained or derived from a recombinant expression system, i.e., from a culture of host cells (e.g., microbial or mammalian) engineered to contain a nucleic acid molecule encoding an IL-7 polypeptide.
  • host cells e.g., microbial or mammalian
  • Microbial refers to recombinant proteins made in bacterial expression systems.
  • the IL-7 conformer is preferably a recombinant human IL-7 conformer.
  • the specific IL-7 conformer has the amino acid sequence of SEQ ID NO:4, 17 or 19 and comprises the disulfide bridges Cys2-Cys92; Cys34-Cys129 and Cys47-Cys141.
  • the specific IL-7 conformer has the amino acid sequence of SEQ ID NO:4, 17 or 19 and comprises an additional Methionine at the N-terminal position (i.e., SEQ ID NO:2).
  • the IL-7 conformer may be a recombinant simian IL-7 conformer.
  • the recombinant simian IL-7 conformer preferably comprises amino acid sequence SEQ ID NO: 13 [SEQ ID NO: 13 comprising or not the signal peptide (amino acids no: 2-25)], SEQ ID NO: 21 or 23.
  • the IL-7 conformer of this invention may be glycosylated or unglycosylated. Many secreted proteins acquire covalently attached carbohydrate units posttranslationally. Glycosylation is frequently in the form of oligosaccharide units linked to proteins through an asparagine residue (situated in the consensus sequence: -Asn-X-Ser/Thr- where X is any amino acid except proline) or through serine/threonine amino acid side chains, giving N-glycosidic or O-glycosidic bonds, respectively. Both the structure and number of oligosaccharide units attached to a particular secreted protein can be highly variable, resulting in a wide range of apparent molecular weights attributable to a single glycoprotein.
  • Murine IL-7 (mIL-7), simian IL-7 (sIL-7) and human IL-7 (hIL-7) are secreted glycoproteins and, in a first variant, the IL-7 conformer of this invention is glycosylated.
  • the conformer may comprise various types of oligosaccharide units, depending on the production system and conditions. These may be, for instance, N-acetyl glucosamine, N-acetyl galactosamine, mannose, galactose, glucose, fucose, xylose, glucuronic acid, iduronic acid and/or sialic acids.
  • the IL-7 conformer of this invention is moderately glycosylated.
  • the glycosylation is of the CHO type, even more preferably, produced by a CHO glycosylation mutant that stably expresses ⁇ 2,6 sialyltransferase and presents a deficiency in CMP-Neu5Ac Hydrolase activity.
  • Such glycosylation typically includes N-acetyl glucosamine, N-acetyi galactosamine, mannose, galactose, glucose, fucose, xylose, glucuronic acid, iduronic acid and/or sialic acids.
  • the IL-7 conformer comprises a human type glycosylation.
  • the IL-7 conformer is typically produced by recombinant technology in a human host cell, which may be selected from human stromal or epithelial cell lines, HEK-293 (Human Embryonic Kidney), HER (Human Embryonic Retina), HEK (Human Epidermal Keratinocytes), human thymus or human cortical epithelial cell lines, human bone marrow or human bone marrow stromal cell lines.
  • the IL-7 conformer is not glycosylated.
  • a recombinant human IL-7 conformer having human carbohydrate pattern is highly preferred for therapeutic use.
  • the invention discloses a specific IL-7 conformer, wherein said IL-7 conformer has the amino acid sequence of SEQ ID NO:4, 17 or 19, comprises the disulfide bridges Cys2-Cys92; Cys34-Cys129 and Cys47- Cys141, and is human glycosylated, moderately glycosylated or unglycosylated, more preferably (human type-)glycosylated.
  • the sequence of the conformer can comprise an additional methionine residue at the N-terminal end (SEQ ID NO:2) and is glycosylated or unglycosylated.
  • the purified conformer may be in the form of a monomer, or associated or complexed with a particular compound of choice.
  • the IL-7 conformer is associated to the hepatocyte growth factor ("HGF"), as a heterodimer.
  • HGF hepatocyte growth factor
  • the heterodimer may be obtained chemically, by complexation or by recombinant technology (i.e., by genetic fusion).
  • the IL-7 conformer can be functionally attached to a Fc portion of an IgG heavy chain, typically through a peptide hinge region. Such fusion molecules have potentially increased stability and half-life in vivo.
  • the IgG moiety is most preferably a human IgG1 or IgG4.
  • the IL-7 conformer can be functionally associated to a human serum albumin ("HSA”) or a portion of a HSA, as a fusion protein.
  • HSA human serum albumin
  • fusion molecules have potentially increased stability and prolonged half-life in vivo.
  • the invention also discloses a drug substance comprising, as the desired product, an IL-7 conformer as described above, said drug substance being substantially free of IL-7 product-related substances and product related impurities.
  • a preferred drug substance is further substantially free of process related impurities.
  • drug substance refers to a product suitable for use as the active principle of a medicament.
  • the "drug substance” according to this invention is, by nature, a complex product, i.e., as a result of its production method (e.g., recombinant DNA technology).
  • an IL-7 drug substance or pharmaceutical composition should contain, as the major molecular species, the conformer refolded according to the 1-4; 2-5; 3-6 disulfide bridges.
  • r-hIL-7 molecular variants such as 1-6; 2-5; 3-4 do exist and have been described in the literature for long term use and/or repeated administrations, the results obtained by the inventors now demonstrate, for the first time, that these products will behave as product related impurities, presenting new epitopes and susceptible to induce a neutralizing immune response toward the preferred conformation of IL-7.
  • the present invention also discloses the fact that, in the specific case of a recombinant IL-7-containing pharmaceutical compositions, other potential product-related substances and product-related impurities, which would normally be included in the specification of the drug substance and/or drug product, although bioactive, behave like new epitopes because of the potent vaccine enhancing property of IL-7. These products should thus be strictly minimized because they are able to trigger an immune reaction against the desired IL-7 molecule.
  • Bioactivity in a specific bioassay is a key critical result to qualify the activity of a recombinant protein for therapeutic use (see ICH Q6B, Harmonized Tripartite Guideline which concerns specifications, test procedures and acceptance criteria for biotechnological drug substances and products. Published in the Federal Register in August 18, 1999; issued as 64FR page 44928 (FDA) and adopted in Europe by the Committee for Proprietary Medicinal Products in March 1999; issued as CPMP/ICH/365/96). Bioactivity is in fact the only single test of activity usually retained as a specification of activity for production batch release. Compliance with bioactivity specifications usually warrants that the recombinant protein drug substance / drug product is active in humans.
  • ICH international guideline Q6B stipulates that " product related substances are molecular variants of the desired product formed during manufacture and / or storage, which are active and have no deleterious effect on the safety and efficacy of the drug product. These variants possess properties comparable to the desired product and are not considered impurities ". In that light, various product related substances and/or product related impurities, molecular variants of IL-7, if bioactive, would authorize a batch release based on the bioassay.
  • the present invention now discloses that a specific attention should be paid to conformers resulting from an inappropriate refolding of the molecule, to deamidated forms, to dimers and to hyperglycosylated forms, for example, and more generally to molecular bioactive variants susceptible to induce an immune reaction because they differ from the preferred, specifically identified, molecular species.
  • the invention thus discloses a drug substance comprising, as the desired product, an IL-7 conformer as described above, said drug substance being substantially free of IL-7 product-related substances and product related impurities.
  • the term "substantially free”, as used herein, indicates that the drug substance contains no significant or adverse amount of product-related substances, product related impurities and process-related impurities. More specifically, the drug substance should contain less than 5%, more preferably less than 3%, even more preferably less than 2% of product-related substances, product related impurities and process-related impurities. Most preferred drug substances contain less than about 1% of product-related substances and/or product related impurities and only trace amount of process-related impurities.
  • IL-7 product-related substances designate IL-7 molecular variants, which include, for example, other IL-7 conformer and/or any active or inactive peptide or polypeptide fragments of IL-7.
  • IL-7-related impurities include, for example, human IL-7 polypeptides comprising mono or bi-disulfide bridges, contiguous disulfide bridges and any disulfide bridges combination such as the following: Cys: 1-6; 2-5; 3-4, and adjacent disulfide bridges: Cys: 1-2; 3-4; 5-6.
  • IL-7 related impurities include aberrant glycosylated form including hyper-glycosylated IL-7, yeast-glycosylated IL-7, truncated IL-7, deamidated recombinant IL-7, dimeric or multimeric protein comprising IL-7, oxidized methionine form or a combination thereof.
  • Process related impurities include DNA, endotoxins, cell debris, etc.
  • the present invention now demonstrates that conventional recombinant methods of producing IL-7 also generate various impurities as described above.
  • the invention more particularly demonstrates that various IL-7 molecular variants are present and that various amino acid sequences are present.
  • the inventors have discovered that the most powerful mid or long term in vivo biological activity of r-IL-7 is carried by a specific conformer having three particular disulfide bridges: Cys: 1-4 (Cys2-Cys92); 2-5 (Cys34-Cys129); 3-6 (Cys47-Cys141).
  • Missing or improper disulfide bridges could very significantly decrease IL-7 activity. Furthermore, the presence of such impurities produces molecules which are immunoreactive upon administration to human or non-human primates, and thus impair the therapeutic benefit of the IL-7 conformer of the present invention.
  • a preferred drug substance is thus a drug substance wherein the total amount by weight of IL-7 comprises at least 95% by weight, preferably at least 98% by weight, more preferably at least 99.5% by weight of an IL-7 conformer according to the invention.
  • the invention also discloses a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a drug substance as described above and one or more pharmaceutically compatible or acceptable carriers, excipients or diluents.
  • the invention shows that pharmaceutical compositions comprising a substantially pure IL-7 conformer as described above clearly increase the vaccine properties of IL-7 and its capacity to stimulate antigen-specific immune responses.
  • the invention thus demonstrates that highly efficient biological preparations are obtained when IL-7 related impurities are removed and a specific conformer as described above is used.
  • the pharmaceutically compatible or physiologically acceptable carrier, excipient or diluent may be selected from neutral to slightly acidic, isotonic, buffered saline, solutions or suspensions and more preferably from sucrose, trehalose, and amino acid.
  • the pharmaceutically compatible carrier is preferably contained in an appropriate buffer to form an isotonic solution.
  • Numerous molecular variants of IL-7 such as hyperglycosylated recombinant proteins produced from recombined yeast strains, IL-7 dimers produced at low ionic strength, and deamidated forms, although bioactive should be strictly minimized. During manufacture and storage, exposition of the molecule to high or moderately high pHs: 8 to 10 triggers the formation of deamidated forms.
  • the appropriate buffer has preferably a pH range comprised between 5 to 7.5, preferably 6 to 7, even more preferably of about 6.5 and is preferably an organic salt selected from a sodium citrate buffer or an ammonium acetate buffer.
  • the pharmaceutical composition may be in the form of a suspension, solution, gel, powder, solid, etc. The composition is preferably a lyophilized form.
  • product can be formulated as a lyophilisate using appropriate excipient solutions (e.g., sucrose and/or trehalose in a range of 1/1 to 10/1, preferably 2/1 to 6/1 sugar/drug substance mass ratio for example) as diluents and lyo-protectors.
  • excipient solutions e.g., sucrose and/or trehalose in a range of 1/1 to 10/1, preferably 2/1 to 6/1 sugar/drug substance mass ratio for example
  • the composition may comprise stabilizing agents, such as sugar, amino acids, proteins, surfactants, etc.
  • the composition may comprise any saline solution, including phosphates, chloride, etc. Appropriate dosages can be determined in trials.
  • a particular pharmaceutical composition can comprise, in addition to the active drug substance, a protein and/or a surfactant.
  • a protein or any other high molecular weight molecule of natural origin, reduces exposition of IL-7 to the host immune system and therefore avoids secondary effects.
  • the protein is non immunogenic in the subject, such as any protein of human origin.
  • a most preferred example of protein is human serum albumin.
  • the surfactant may be selected from known surfactants such as Tween products, preferably Tween 80.
  • a specific composition of this invention comprises human serum albumin (preferably 2 to 5 mg/ml) or Tween 80 (typically 0.005%) or any other substance such as a tensioactive substance, capable of preventing IL-7 immunogenicity due to local persistence of the drug product after administration of the composition.
  • compositions of the present invention may be adapted so as to obtain an amount of active ingredient that is effective to obtain a desired therapeutic response for a particular composition and method of administration.
  • the selected dosage level therefore depends upon the desired therapeutic effect, the route of administration, the desired duration of treatment and other factors.
  • dosages of 3 to 300 ⁇ g/kg , preferably, 10 to 100 ⁇ g/kg , administered by subcutaneous route can be expected to induce a biological effect preferably while administered from once daily down to once weekly, possibly twice weekly, preferably not more frequently than once every 48h.
  • the specific dose level for any particular patient will depend upon a variety of factors including the body weight, general health, sex, diet, time and route of administration, rates of absorption and excretion, combination with other drugs and the severity of the particular disease being treated.
  • IL-7 a particular attention should be paid to the immune status of the patient before adjusting dose level. The more the patient is immuno-depressed, as judged for instance through peripheral CD4 T-Cell counts, the less the dose necessary to induce a relative increase in lymphocyte counts.
  • recombinant IL-7 has a surprisingly long blood half life in primates. Its effect on cell cycling lasts for 24 to 48 hours, allowing efficient treatment schedules with single injection daily down to one injection weekly.
  • compositions according to the invention are preferably administered from once daily down to once weekly, possibly twice weekly, preferably not more frequently than once every 48h, only in order to obtain and/or stimulate patients immune regeneration.
  • parenteral routes are parenteral routes.
  • the parenteral route is preferably an intra-tumoral, more preferably an intra-venous or a snub-cutaneous administration. It includes also intra-arterial, intra-peritoneal or intra-muscular injections. It should be understood, however, that any other suitable administration route may be contemplated depending upon the health status and the reactivity of the patient.
  • the pharmaceutical composition may comprise additional active ingredients, such as immuno-stimulating agents, preferably selected from a hematopoietic cell growth factor, a cytokine, an antigenic molecule (or antigen) and an adjuvant, for combined, separate or sequential use.
  • immuno-stimulating agents preferably selected from a hematopoietic cell growth factor, a cytokine, an antigenic molecule (or antigen) and an adjuvant, for combined, separate or sequential use.
  • Such additional active ingredients may be formulated in combination with the IL-7, or, separately, for combined, separate or sequential use.
  • the active ingredients are formulated together, in the same recipient or vessel.
  • they are conditioned separately, i.e., in distinct vessels or recipients.
  • the ingredients may be administered separately, e.g., simultaneously or sequentially (e.g., at different injection sites or at different time points), to produce the most efficient biological effect.
  • repeated administrations of one or the two active ingredients may be performed.
  • the invention discloses a pharmaceutical composition
  • a pharmaceutical composition comprising a substantially pure IL-7 conformer as described above and an active ingredient selected from an immuno-stimulant and an antigenic molecule, for combined, separate or sequential use.
  • Adjuvants are preferably formulated separately.
  • the hematopoietic cell growth factor is preferably selected from the Stem Cell Factor (SCF), particularly the soluble form of the SCF, G-CSF, GM-CSF, Flt-3 ligand, IL-15 and IL-2.
  • SCF Stem Cell Factor
  • cytokines for vaccine enhancement include cytokines that induce and/or stimulate a Th1-type immune response.
  • the cytokine is preferably selected from ⁇ interferon, IL-2, IL-12, RANTES, B7-1, MIP-2 and MIP-1 ⁇ . It should be understood that other factors such as FGF7 or FGF10, interleukins and/or hormones may be used in combination with IL-7 to provide additional therapeutic benefit.
  • a specific composition can comprise a substantially pure IL-7 conformer as described above and Stem Cell Factor, particularly the soluble form thereof, IL-15 and/or Flt-3 ligand and/or FGF10.
  • An other specific composition can comprise a substantially pure IL-7 conformer as described above and a cytokine selected from ⁇ interferon, IL-2, IL-12, RANTES and MIP-1a.
  • An other specific composition can comprise a substantially pure IL-7 conformer as described above, a Stem Cell Factor and a cytokine.
  • antigens include antigens derived from HIV, Varicella Zoster virus, Influenza virus, Epstein Barr virus, type I or 2 Herpes Simplex virus, human cytomegalovirus, Dengue virus, Hepatite A, B, C or E virus, Syncytium respiratory virus, human papilloma virus, mycobacterium tuberculosis, Toxoplasma and Chlamydia.
  • composition comprising a substantially pure IL-7 conformer as described above and an antigenic molecule, for combined, separate or sequential use.
  • the composition may further comprise one or several immuno-stimulating agents as disclosed above, for combined, separate or sequential use.
  • compositions comprising an IL-7 conformer as described above, wherein said pharmaceutical composition is administered simultaneously, a few days before or sequentially with one or several antigenic molecules in order to obtain and/or stimulate an antigen-specific immune response in a subject.
  • a method of causing or enhancing an antigen-specific immune response in a subject comprising administering to a subject said antigen (or an epitope-containing fragment thereof) and a pharmaceutical composition comprising an IL-7 conformer as described above.
  • the pharmaceutical composition may be administered simultaneously, a few days before or sequentially with said antigen in order to obtain and/or stimulate an antigen-specific immune response in a subject.
  • the composition may further comprise an adjuvant.
  • the adjuvant may be selected from any substance, mixture, solute or composition facilitating or increasing the immunogenicity of an antigen and able to induce a Th1-type immune response, such as CpG, QS21, ISCOM and monophosphoryl lipid A. Such adjuvants are particularly suited to produce and/or amplify a specific immune response against antigen(s) in mammalian subjects, particularly in humans.
  • the adjuvant is preferably conditioned and administered separately from the IL-7 containing composition and/or at a distinct site of injection, preferably with the desired antigen(s).
  • the present invention also discloses a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a human IL-7 conformer according to the invention in admixture with a suitable diluent, excipient or carrier, for parenteral administration to a human patient for prophylactic or therapeutic stimulation of B or T lymphocyte development and proliferation, or for augmentation of an immune response.
  • the pharmaceutical compositions induce a prolonged lymphopoiesis stimulation and/or amplified immune responses.
  • a pharmaceutical composition according to the invention may also be used in a human patient for prophylactic or therapeutic stimulation of B or T lymphocyte development and proliferation, for enhancement of global and/or specific immuno-reconstitution, or for enhancement of humoral and/or cellular immune responses.
  • a particular pharmaceutical composition according to the invention is for use to prevent or reduce opportunistic infections in immunodeficient patients.
  • Another particular pharmaceutical composition according to the invention is for use to prolong lymphopoiesis stimulation and/or to produce specific immune response not only against dominant epitopes but also against sub-dominant or less immunogenic epitopes, epitopes having a lower affinity for the T cell receptor, which will allow to broaden the repertoire of a specific immune response in human patients.
  • the invention is particularly suited to produce a preventive or curative immune response in subjects, such as immunodeficient patients, cancer patients, patients undergoing grafts, patients infected with a virus or a parasite, elderly patients or any patients having low CD4 count etc.
  • An other aspect of the present invention is to disclose appropriate constructs and methods for producing the above compositions, particularly the above IL-7 conformer and drug substances, in sufficient quantities and quality for pharmaceutical use thereof.
  • nucleic acid molecules comprising a sequence encoding an IL-7 polypeptide, said sequence being optimized for the expression, in a recombinant host, of biologically active IL-7, particularly of an IL-7 conformer as described above. More specifically, the nucleic acid molecules provide for a limited expression of truncated IL-7 polypeptides and increase the yields of production of biologically active human IL-7 conformer.
  • the human and simian IL-7-encoding DNA sequence (genomic or cDNA) contain, at position 49 after the "ATG” initiation codon, a second putative "ATG” which could behave as a second initiation codon in E. Coli. Indeed, this second "ATG” is preceded by a "pseudo Shine-Dalgamo" sequence (ribosome binding sequence).
  • this putative second initiation codon thereby significantly reducing the potential production of an amino-terminal truncated form of r-hIL-7.
  • nucleotides in the coding sequence were mutated in order to inactivate the "SD-like" sequence, without modifying the resulting, encoded, r-IL-7 amino acid sequence.
  • nucleic acid molecules encoding a human IL-7 polypeptide, wherein said nucleic acid contains a modified sequence avoiding the production of an amino-terminal truncated polypeptide, more specifically by inactivation of the "SD-like" sequence (i.e., of the putative second initiation codon located at position 49 in the sequence, which encodes methionine residue 18 in SEQ ID NO: 2).
  • Inactivation of the putative second initiation codon may be achieved, more preferably, by modifying the sequence in at least one of the 5 codons preceding the ATG codon encoding methionine 18 in SEQ ID NO: 1, i.e., in codons 13-17 encoding amino acid sequence TyrGluSerValLeu.
  • the sequence is modified by altering the codon coding for a serine at position 15 in SEQ ID NO: 1 or 2. More specifically, in a preferred sequence, this codon is modified so as not to contain an AG doublet. Suitable codons may be selected, for instance, from TCC, TCT, TCA and TCG. In SEQ ID Nos: 1, 3, 16 and 18, the TCC codon has been used.
  • the codon encoding Tyr at position 13 may be selected from TAC or TAT.
  • the codon encoding Glu at position 14 may be selected from GAG and GAA.
  • the codon encoding Val at position 16 (based on the numbering in SEQ ID NO:1) may be selected from GTT, GTC, GTA and GTG.
  • the codon encoding Leu at position 17 (based on the numbering in SEQ ID NO:1) may be selected from CTG, CTA, CTT, CTC, TTA and TTG. All combinations of the above indicated codons are possible.
  • a specific example of an altered DS-like sequence corresponds to nucleotides 37-51 of SEQ ID NO:1.
  • a specific, preferred nucleic acid molecule comprises the sequence of SEQ ID NO:1 and codes for a recombinant human IL-7 polypeptide.
  • nucleic acid molecule as described above which further comprises a signal sequence causing secretion of the produced polypeptide.
  • the signal sequence may be selected from the natural signal sequence of the human IL-7 protein, or from any heterologous signal sequence. Such sequences may originate from other secreted proteins, such as the human growth hormone, or be artificial or synthetic.
  • a preferred signal peptide may be a natural signal peptide of a human growth factor or of a human growth hormone and, more preferably, a natural signal peptide of the human erythropoietin.
  • An even more preferred signal peptide is a synthetic signal peptide such as HMM38.
  • Preferred sequences are those functional in competent mammalian host cells. A specific example of such an improved sequence comprises SEQ ID NO:3, 16 or 18. This sequence comprises a leader sequence and an inactivated SD-like sequence, for improved expression.
  • polypeptide encoded by a nucleic acid sequence as described above, which may be glycosylated or unglycosylated.
  • polypeptide as described above, wherein the tertiary structure of said polypeptide comprises the following three disulfide bridges: Cys: 1-4; 2-5; 3-6.
  • the present invention further discloses vectors comprising a nucleic acid as described above, as well as recombinant host cells comprising said nucleic acid or said vectors.
  • the nucleic acids and vectors may be used to produce recombinant human IL-7 polypeptides in various competent host cells, as well as for gene therapy purposes.
  • the vector may be a plasmid, virus, phage, cosmid, episome, etc.
  • Preferred vectors are viral vectors (e.g., recombinant adenoviruses) and plasmids, which can be produced based on commercially available backbones, such as pBR, pcDNA, pUC, pET, pVITRO, etc.
  • the vector typically comprises regulatory elements or sequences to control or mediate expression of an IL-7 polypeptide from the optimized coding nucleic acid.
  • the regulatory sequences may be chosen from promoters, enhancers, silencers, tissue-specific signals, peptide signals, introns, terminators, polyA sequences, GC regions, etc., or a combination thereof.
  • Such regulatory elements or sequences may be derived from mammalian, plant, bacterial, yeast, bacteriophage or viral genes, or from artificial sources.
  • Useful promoters for prokaryote expression include T7 RNA polymerase promoter (pT7), TAC promoter (pTAC), Trp promoter, Lac promoter, Tre promoter, PhoA promoter for example.
  • Suitable promoters for expression in mammalian cells include viral promoters (e.g., CMV, LTR, RSV, SV40, TK, pCAG, etc.), domestic gene promoters (e.g., E1f ⁇ , chicken ⁇ actine, Ubiquitine, INSM1, etc.), hybrid promoters (e.g., actine / globin, etc.), etc.
  • a vector may comprise more than one promoter.
  • the promoters may be inducible or regulated. For instance, the use of inducible or regulated promoters allows a better control of production by dissociating the culture from production phases.
  • Inducible or regulated promoters may be found in the literature, such as the Tetracycline system, the Geneswitch system, the Ecdysone system, the Oestradiol system, the RU486 system, the Cumate system, the methallothioneine promoter etc.
  • Other systems are based on electric currents or microwaves, such as Ultra sons focalises and the like. These systems can be used to control expression of an IL-7 polypeptide according to the invention.
  • the IL-7 may be co-expressed with an anti-apoptotic factor (e.g., iex, Bcl2, BclXL, etc.).
  • an anti-apoptotic factor e.g., iex, Bcl2, BclXL, etc.
  • the cDNA coding for said IL-7 and for said anti-apoptotic factor
  • the cDNA may be both placed downstream of the same promoter, but separated by an IRES sequence, or each of them downstream of its own promoter.
  • the vector may further comprise an origin of replication and/or a marker gene, which may be selected from conventional sequences.
  • the vector may further comprise various combinations of these different elements which may be organized in different ways.
  • the present invention thus also discloses recombinant host cells comprising a nucleic acid or a vector as described above.
  • the host cell may be selected from any eukaryotic and prokaryotic cells, typically from a mammalian cell (in particular a human, rodent, canine cell), a bacterial cell (in particular E. coli, Bacillus Brevis, Bacillus Subtilis), a yeast cell, a plant cell and an insect cell. Where yeast, plant or insect cells are used for production purposes, the products obtained in said cells are preferably submitted to a treatment step in order to produce unglycosylated products.
  • These host cells may be adapted to serum-free media. Production may also be accomplished in a transgenic animal.
  • Preferred recombinant host cells are selected from a mammalian cell, in particular a human cell, and a bacterial cell, as well as derivatives or mutants thereof.
  • suitable host cells include bacteria, which provide for non-glycosylated production of proteins.
  • Bacteria e.g., Escherichia coli
  • recombinant IL-7 is generally expressed as inclusion bodies. This host is particularly advantageous and may be used, more specifically, with a vector comprising a sequence SEQ ID NO:1.
  • Other examples of suitable host cells include mammalian cells, which provide for glycosylated production of proteins.
  • IL-7 may be produced as a secreted protein using functional signal peptide sequences.
  • These hosts may be used, more specifically, with a vector comprising a sequence SEQ ID NO:3, 16 or 18.
  • a prokaryotic host cell comprising a nucleic acid molecule comprising SEQ ID NO: 1.
  • a eukaryotic host cell comprising a nucleic acid molecule comprising SEQ ID NO: 3,12, 16, 18, 20 or 22.
  • a eukaryotic or prokaryotic host cell comprising a nucleic acid molecule encoding an IL-7 polypeptide comprising amino acids 26-152 of SEQ ID NO: 13.
  • antibodies immunoreactive with an IL-7 conformer as described above.
  • Such antibodies may be produced according to conventional methods, including immunization of animals and collecting the serum (polyclonal) or preparing hybridomas from spleen cells (monoclonal).
  • Fragments e.g., Fab'
  • derivatives of antibodies e.g., ScFv
  • Preferred antibodies are specifically immunoreactive with an IL-7 conformer as described above, i.e., can bind the IL-7 conformer without substantially binding IL-7 polypeptides which do not comprise the following three di-sulfide bridges: Cys2-Cys92, Cys34-Cys129 and Cys47-Cys141.
  • non-specific or less effective binding to such other antigens may be observed, such non-specific binding can be distinguished from specific binding to the particular conformer of this invention.
  • the antibody is preferably of simian, murine or human origin or has been humanized.
  • the invention also relates to a hybridoma cell line that produces a monoclonal antibody as described above.
  • Such antibodies are useful in detecting IL-7 conformer or in neutralizing IL-7 biological activity in assays or experiments involving multiple lymphokines.
  • a composition suitable for diagnosis, assay or therapy comprising such monoclonal antibodies is also an object of the present invention.
  • the invention also discloses a method of producing or amplifying an optimized IL-7 nucleic acid from a sample, the method comprising:
  • the sample may comprise genomic DNA, cDNA or RNA.
  • Amplification of the gene may be produced by any technique known per se, such as PCR, RT-PCR, or other amplification techniques. Amplification usually requires the use of a pair of primers characterized in that the forward sequence hybridizes with the 5' end of the DNA and in that the reverse sequence hybridizes with the 3' end of the same DNA. Specific examples of primers are as follows :
  • primers can be designed by the skilled artisan, such as fragment of the optimized IL-7 gene, for use in the amplification step and especially a pair of primers comprising a forward sequence and a reverse sequence, wherein said primers of said pair hybridize with a region of the mutated IL-7 gene and allow amplification of at least a portion of the IL-7 gene.
  • Another object of the present invention discloses processes which can be used, on an industrial scale, for the production of a pharmaceutical grade, substantially pure IL-7 conformer as described above.
  • the process leads to high yields of recombinant IL-7 conformer suitable for therapeutic use.
  • the invention also discloses novel methods of controlling IL-7-containing compositions, to determine the presence of amount of an IL-7 conformer as described above.
  • the method of producing an IL-7 conformer or a drug substance as defined above comprises:
  • the sample used in step a) may be any biological sample containing an IL-7 polypeptide.
  • Preferred samples are cell supernatants of recombinant host cells that produce a recombinant IL-7 polypeptide, more preferably a recombinant human or simian IL-7 polypeptide, even more preferably, a recombinant human IL-7 polypeptide.
  • Most preferred samples are (obtained from) cell supernatants of recombinant host cells that contain an optimized IL-7 coding nucleic acid molecule as described above, more particularly a polynucleotide comprising SEQ ID NO: 1, 3, 12, 16, 18, 20 or 22.
  • the sample may be subjected to various treatments or conditions in order to increase purity of IL-7, to release IL-7 from inclusion bodies, to remove cell debris or other particular bodies, etc. Typical examples of such treatments include centrifugation, filtration and/or clarification.
  • the sample may thus be enriched for IL-7 polypeptide.
  • the sample is (or derives from) a culture of prokaryotic host cells encoding an IL-7 polypeptide
  • treatments of the sample are necessary. Such treatments include:
  • denaturing conditions may be applied as well, alone or in combination.
  • a preferred level of denaturation corresponds to the destruction of all the disulfide bridges of IL-7.
  • a preferred full denaturation protocol uses 8 M guanidine hydrochloride buffer. High guanidine hydrochloride molarity is preferred to allow full denaturation of the protein prior to its correct refolding. This stage of denaturation may be controlled by analytical techniques known in the art, such as electrophoresis, for instance.
  • step ii) may be performed by different techniques known per se, but which have not been used so far in the present combination to produce a fully active IL-7 polypeptide conformer. These techniques are more preferably selected from hydrophobic interaction chromatography, ion exchange chromatography, affinity chromatography and gel filtration chromatography, either alone or in various combinations. Such methods allow to remove DNA and other impurities (lipids, etc.) which would lower recovery of the subsequent refolding step.
  • step ii) comprises a hydrophobic interaction chromatography step. Such chromatography may be carried out using various supports and formats, preferably using HIC butyl, preferably in denaturing conditions. To perform this step, a preferred pH range is comprised between 6 and 9, preferably between 7 and 8,5 inclusive. Step ii) may be carried out on any support, preferably on batch or in column using an appropriate gel.
  • the refolding step iii) comprises a renaturation and preferably reoxydation of the IL-7 polypeptides and, typically, a further desalting step performed by filtration (for example a gel filtration) or ultrafiltration.
  • the refolding comprises an ion exchange chromotography step to separate the active new conformer from the unwanted forms.
  • the refolding comprises a step of contacting the purified polypeptides with a selected affinity chromatography support in order to separate the active new conformer from the unwanted forms. This very advantageous step is selective for the elution of the correctly refolded IL-7.
  • the refolding step iii) comprises passing the solution obtained in step ii) through a column comprising polymers of sulfated polysaccharides.
  • the sulfated polysaccharide is preferably dextran sulfate or heparin and the matrices can be sepharose, acrylamide, agarose, dextran, cellulose or other types commonly employer in protein purification.
  • a preferred matrix is heparin sepharose.
  • the process may further comprise an additional step of eluting the desired IL-7 conformer.
  • affinity column may be used both to purify and refold the polypeptides, i.e., to perform simultaneously steps ii) and iii).
  • the sample is (or derives from) a culture of eukaryotic host cells encoding IL-7 polypeptides
  • the sample may not be necessary to perform the above treatment steps i) to iii).
  • the sample may comprise correctly folded IL-7 proteins, although in complex admixture with other product-related substances and impurities.
  • the recombinant eukaryotic host cell comprises a nucleic acid molecule causing expression and secretion of the IL-7 polypeptides in the culture medium.
  • the sample may be directly subjected to the purification step b).
  • Purification step b) may comprise one or several purification steps such as filtration or ultrafiltration steps ion exchange chromatography, , affinity chromatography, hydrophobic interaction chromatography etc., in order to eliminate drug related substances such as other conformers, deamidated forms, dimers of the protein, residual impurities, including DNA, endotoxins, etc.
  • the chromatography step may be realized in a flow through mode, in a capture mode or in a streamline mode.
  • the purification step b) can comprise loading the sample through a column packed with a specific gel comprising sulfated polysaccharides immobilized on a resin (dextran sulfate or heparin for example).
  • the purifying step can comprise loading the sample through a column packed with a specific gel comprising a monoclonal anti IL-7 antibody immobilized on a resin (dextran sulfate or heparin for example).
  • IL-7 conformer allows the reproducible and efficient production of a substantially pure fully active human IL-7 conformer as described above.
  • the methods are particularly advantageous since the recombinant IL-7 conformer can be obtained with a purity of at least 95% by weight, preferably at least 98% by weight and even more preferably at least 99% or even 99.5% by weight with respect to the total amount of IL-7.
  • the above purification method may be applied to polypeptides other than human IL-7, particularly to other cytokines having one or several disulfide bridges, including animal IL-7 (e.g., monkey IL-7), IL-13, IL-15, etc.
  • the invention discloses a method of producing an IL-7 conformer as defined above, the method comprising at least the following steps:
  • the invention discloses a method of producing an IL-7 conformer as defined above, the method comprising at least the following steps:
  • IL-7 expression by the recombinant host cells is inducible, regulated or transient, so that the cell culture and IL-7 expression phases can be dissociated. More particularly, in a particular embodiment, IL-7 expression can be repressed or minimized during recombinant cell growth, expansion and/or culturing, to allow the production of large amounts of recombinant host cells without any IL-7-mediated potential toxic effect. Then, IL-7 expression can be induced within the cell culture (or on a sample thereof), allowing the efficient synthesis and release of recombinant IL-7.
  • An object of this invention thus also discloses a method of producing a recombinant IL-7 polypeptide, comprising culturing a recombinant host cell comprising a nucleic acid molecule encoding said IL-7 polypeptide and recovering the recombinant IL-7 polypeptide produced, wherein said nucleic acid molecule provides for a regulated or inducible expression of said IL-7 polypeptide, so that expression of said IL-7 polypeptide can be repressed or minimized during recombinant cell growth and induced during production phase.
  • the nucleic acid typically comprises an inducible promoter, which can be repressed or activated in the presence or absence of a specific agent contained or added into the culture media. The method is particularly suited to produce an IL-7 conformer as disclosed above.
  • regulated or inducible expression systems may be used in various cells, such as for instance HEK293, HEK293 EBNA, HEK, T-REX TM -293, T-REX TM -HeLa, T-REX TM -CHO or T-REX TM -Jurkat cell lines, transformed with a recombinant vector designed to express recombinant IL-7 after induction.
  • transient transfection can be used to dissociate cell expansion from IL-7 production.
  • efficient gene delivery vectors are used to introduce an IL-7 coding sequence into cells upon expansion thereof.
  • the vector system for transient transfection is a viral vector, such as a recombinant adenovirus or an episomal vector [e.g., pCEPH (Invitrogene), pTT (IRB: Durocher Y. et al. Nucl. Acids. Res., 2002, 30(2 )) or using MAR sequences].
  • Adenoviruses and other viral vectors such as AAVs, for instance
  • E1-defective adenoviruses are produced in a E1-complementing cell line, such as HEK293, PERC6 cells, etc.
  • a E1-complementing cell line such as HEK293, PERC6 cells, etc.
  • Such transient transfection process can be implemented in various mammalian cells in culture, such as A549, HeLa, VERO, BHK or CHO transformed cells for example (as disclosed in example A4).
  • An alternative transient expression method suitable for use in the present invention is disclosed for instance in the next article: Durocher Y. et al. Nucl. Acids. Res., 2002, 30(2 ) in HEK293 EBNA or HEK293 cells.
  • the invention also discloses a method to identify and/or measure the quantity of IL-7 conformer and/or related impurities in a sample, particularly in a pharmaceutical preparation.
  • characterization methods can be used to initially characterize and qualify the protein for filing a therapeutic use, in quality control of pharmaceutical batches.
  • the present invention now demonstrates that a fully biologically active IL-7 conformer exists and that the presence of impurities such as other conformers or IL7 peptide fragments may generate highly undesirable side effects.
  • the invention thus proposes, for the first time, a method of characterizing and controlling IL-7-containing preparations, to determine the presence and/or relative quantity of the specific IL-7 conformer of this invention.
  • Preferred methods use western blot, size-exclusion HPLC, Escherichia Coli Protein (ECP) assay, bacterial endotoxins assay, Limulus Ameobocyte Lysate test (LAL test), DNA quantification, SDS-PAGE, reverse phase HPLC, ion exchange HPLC, heparin Affinity HPLC, Bicinchoninic Acid Assay (BCA) method, Amino Acid Assay (AAA) method, ELISA, UV absorption and/or a Bioassay. These methods may be carried out alone or in various combinations.
  • the step is particularly suited for quality control of clinical or pharmaceutical compositions, whereby only compositions comprising more than about 95% of the above IL-7 conformer are retained, preferably more than about 96%, 98% or 99,5%.
  • Disclosed is also the use of the isolated IL-7 polypeptide conformer or of a recombinant IL-7 conformer obtained with the processes as described above, for the manufacture of a pharmaceutical composition to prevent or treat a disease associated with an immunodeficiency, particularly to induce a prolonged lymphopoiesis stimulation, to cause and/or amplify an immune response, particularly an antigen-specific immune response.
  • IL-7 conformer Disclosed is also the use of an IL-7 conformer as a tool for experimental and pharmacological use in monkeys.
  • kits for implementing the above methods comprising at least a primer specific for the optimized IL-7 gene and, optionally, a pair of primers and/or a probe and/or reagents for a nucleic acid amplification reaction and/or antibodies, as described above.
  • Kits may alternatively comprise reagents to produce an IL-7 polypeptide as described above, such as an optimized nucleic acid, a vector, a recombinant host cell, and/or protocols or reagents for the purification or quality control of such preparations.
  • the human IL-7 encoding cDNA sequence was amplified by polymerase chain reaction (PCR) ( Mullis et al.; 1987; Methods in Enzymology; 155:335-350 ) from human placenta cDNA (BioChain Inc.) using the following specific oligonucleotide primers which contain restriction endonuclease recognition sequences:
  • the human IL-7-encoding DNA sequence presents, at position 49 after the "ATG” initiation codon, a second putative "ATG” which could behave as a second initiation codon in E. Coli, since this second "ATG” is preceded by a "pseudo Shine-Dalgamo" sequence (ribosome binding sequence).
  • a second putative "ATG” which could behave as a second initiation codon in E. Coli, since this second "ATG” is preceded by a "pseudo Shine-Dalgamo” sequence (ribosome binding sequence).
  • a "pseudo Shine-Dalgamo" sequence ribosome binding sequence
  • the suppression of the SD-like sequence in the IL-7-encoding DNA sequence was performed by site directed mutagenesis PCR, using the following oligonucleotide primers:
  • the amplification and suppression of the SD-like sequence may be realized by single step using oligonucleotide primers above.
  • PCR products were assayed by polyacrylamide or Agarose gel electrophoresis in the presence of ethidium bromide and visualization by fluorescence of DNA bands stimulated by UV irradiation.
  • a single major product band of the size corresponding to the IL-7 PCR fragment was isolated and inserted into the plasmid vector pCR II-TOPO (Invitrogen) using the TA-cloning method.
  • the ligation products were transformed into TOP10 competent cells (Invitrogen).
  • Plasmid Miniprep Isolation techniques Biorad
  • dideoxy Sequencing Sanger et al.; 1977; Proceedings of the National Academy of Sciences of the USA; 74:5463-5467 ) of an asymmetric PCR product DNA using pCR II TOPO universal primers (Invitrogen) as sequencing primers.
  • plasmid DNA from a positive clone was digested with restriction endonucleases BamHI and Ndel and the resulting fragment, r-hIL-7 encoding DNA sequence, was inserted in between BamH I and Nde I restriction sites of a ptac vector.
  • This expression plasmid was constructed by replacing the T7 promoter by a tac promoter in a classical pET vector derived from pBR322.
  • the tac promoter was first amplified using pMAL-p2X (Biolabs) as a matrix and oligonucleotides :
  • the obtained PCR fragment was loaded on an agarose gel to check for its correct size. It was then digested by the B gl II and X ba I and inserted into the pET9a (Novagen) hydrolyzed by the same enzymes. The obtained ligation products were transformed into TOP10 (Invitrogen) competent cells and selected on their kanamycine resistance. The obtained ptac vector was then checked by digestion with several enzymes and by sequencing analysis using oligonucleotides ptac1 and ptac2 as sequencing primers.
  • E. Coli final expression plasmid comprising SEQ ID NO:1, called ptac-hIL-7 (cf. : Figure n°1), was subcloned into E. coli JM101 cells (ATCC).
  • the simian (Macaca Mulatta Rhesus monkey) IL-7 encoding cDNA sequence was obtained from rhesus monkey kidney cDNA (BioChain Inc.) using PCR amplification ( Mullis et al.; 1987; Methods in Enzymology; 155:335-350 ).
  • the basic strategy was to amplify simian IL-7 cDNA by PCR with specific oligonucleotide primers used to amplify human IL-7 cDNA (SEQ ID NO: 5: IL75' and SEQ ID NO: 6: IL73').
  • SEQ ID NO: 5: IL75' and SEQ ID NO: 6: IL73' A single band was apparent on gel electrophoresis (cf. Figure 6 ).
  • the simian IL-7-encoding DNA sequence presents at position 49 after the "ATG” initiation codon, a second putative "ATG” which could behave as a second initiation codon in E. Coli , since this second "ATG” is preceded by a "pseudo Shine-Dalgamo” sequence (ribosome binding sequence).
  • the amplification of the simian IL-7 encoding DNA sequence lacked of the SD-like sequence was performed by site directed mutagenesis PCR, using oligonucleotide primers: SEQ ID NO: 7: mutIL75' and SEQ ID NO: 6: IL73').
  • PCR products were analyzed by polyacrylamide or agarose gel electrophoresis in the presence of ethidium bromide and visualization by fluorescence of DNA bands stimulated by UV irradiation.
  • a single major product band of the size corresponding to the IL-7 PCR fragment (cf. Figure 7 ) was isolated and inserted into the plasmid vector pCR II-TOPO (Invitrogen) using the TA-cloning method.
  • the ligation products were transformed into TOP10 competent cells.
  • Plasmid Miniprep Isolation techniques To select positive clones, plasmid DNA, prepared from cultured individual bacterial clones kanamycin resistant by Plasmid Miniprep Isolation techniques, were analyzed by restriction mapping and confirmed by dideoxy Sequencing ( Sanger et al.; 1977; Proceedings of the National Academy of Sciences of the USA; 74:5463-5467 ) of an asymmetric PCR product DNA using pCR II TOPO universal primers (Invitrogen) as sequencing primers yielded about 700 bases containing sIL-7 sequences.
  • Plasmid DNA from a positive clone was digested with restriction endonucleases BamHI and Ndel and the resulting fragment, r-sIL-7 encoding DNA sequence, was inserted into ptac vector, as descibed in example 1.1., which was also digested with BamH I and Nde I restriction sites. The ligation products were transformed into TOP10 competent cells. The selection for plasmid-containing cells was on the basis of the antibiotic (kanamycin) resistance marker gene carried on the vector. Plasmid DNA from a positive clone was isolated from cultured cells, selected by restriction mapping and confirmed by sequencing analysis using T7 terminator universal primer on one hand and ptac promoter primer on the other hand as sequencing primers.
  • E. Coli final expression plasmid comprising SEQ ID NO: 12, called ptac-sIL7opt (cf. : Figure n°2), was subcloned into E. coli JM101 cells.
  • the human IL-7 encoding cDNA sequence was amplified by polymerase chain reaction (PCR) ( Mullis et al.; 1987; Methods in Enzymology; 155:, 335-350 ) from human placenta cDNA (BioChain Inc.), using oligonucleotides as primers in attempts to amplify the portions of the cDNA of interest: the IL-7 cDNA fragments linked at the 5' end to the natural IL-7 peptide signal.
  • PCR polymerase chain reaction
  • oligonucleotides which contain restriction endonuclease recognition sequences and kozak sequence:
  • PCR products were assayed by polyacrylamide or agarose gel electrophoresis in the presence of ethidium bromide and visualization by fluorescence of DNA bands stimulated by UV irradiation (cf. figure 8 ).
  • the product band of the size corresponding to the IL-7 PCR fragment, designated "hPSIL-7 cDNA” was isolated and inserted into the plasmid vector pCR II-TOPO (Invitrogen) using the TA-cloning method.
  • the ligation products were transformed into TOP10 competent cells.
  • plasmid DNA prepared from cultured individual bacterial clones ampicilin resistant by Plasmid Miniprep Isolation techniques (Biorad), were analyzed, by restriction mapping and confirmed by dideoxy sequencing ( Sanger et al.; 1977; Proceedings of the National Academy of Sciences of the USA; 74:5463-5467 ) of an asymmetric PCR product DNA using pCR II TOPO universal primers as sequencing primers.
  • Plasmid DNA from a positive clone was digested with restriction endonucleases HindIII and BamHI and the resulting fragment, "hPSIL-7 cDNA" fragment, was inserted into pcDNA3.1(+) vector polylinker (Invitrogen) which had been digested with BamH I and HindIII restriction sites.
  • This vector includes a gene for the selection and amplification of clones.
  • the ligation products were transformed into TOP10 competent cells. The selection for plasmid-containing cells was on the basis of the antibiotic (ampicilin) resistance marker gene carried on the vector.
  • Plasmid DNA from a positive clone was isolated from cultured cells, selected by restriction mapping and confirmed by sequencing analysis using pcDNA3.1 (+) universal primers as sequencing primers (Invitrogen).
  • the expression system was designed to express an IL-7 protein predicted from the translation of the natural human IL-7 gene sequence. Selection for recombinant vector-containing cells was done on the basis of the antibiotic (Ampicilin for cloning in E. Coli and Neomycin for expression in mammalian cells) resistance marker gene carried on the vector.
  • the mammalian (HEK-293, CHO or BHK) expression vector comprising SEQ ID NO:3, is called pcDNA-hPSIL-7 (cf.: Figure n°3). Expression of human IL-7 in HEK-293 or CHO transfected cells was achieved using the expression vector pcDNA-hPSIL-7. After linearization by B gl II, expression vector, pcDNA-hPSIL-7, was transfected in the mammalian host cells using methods known to those skilled in the art. The selectable marker used to establish stable transformants was G418 (Invitrogen).
  • the human IL-7 encoding cDNA sequence was isolated from the pcDNA-hPSIL-7 by digestion with restriction endonucleases Hind III and BamH I.
  • the resulting "hPSIL-7 cDNA" fragment was purified on agarose gel and inserted into the pcDNA4/TO (Invitrogen) hydrolyzed with the same enzymes.
  • the ligation products were transformed into TOP10F' competent cells (Invitrogen).
  • the selection for plasmid-containing cells was on the basis of the antibiotic (Ampicillin) resistance marker gene carried on the vector. Plasmid DNA from positives clones were isolated from cultured cells, checked by restriction mapping and confirmed by sequencing analysis using the universal CMV forward and BGH reverse primers.
  • the expression system was designed to express, after induction with tetracycline or analogous, an IL-7 protein predicted from the translation of the natural human IL-7 gene sequence. Selection for recombinant vector-containing cells was done on the basis of the antibiotic (Ampicillin for cloning in E. coli and Zeocin for expression in mammalian cells) resistance marker gene carried on the vector).
  • the inducible mammalian expression vector is called phT-PSIL7h (cf.: Figure n°14).
  • Expression of human IL-7 in HEK-293, HEK, T-RE) TM -293, T-REX TM -HeLa, T-REX TM -CHO or T-REX TM -Jurkat transfected cells was achieved using the expression vector phT-PSIL7h. After linearization by Pvul, expression vector, phT-PSIL7h, was transfected in the mammalian host cells using methods known to those skilled in the art. The selectable marker used to establish stable transformants was zeocin (Invitrogen).
  • the human IL-7 encoding cDNA sequence was amplified by polymerase chain reaction (PCR) ( Mullis et al.; 1987; Methods in Enzymology; 155:335-350 ) from the pcDNA-hPSIL-7, using oligonucleotides as primers in attempts to amplify the cDNA of interest (IL-7 cDNA linked to its natural peptide signal) between EcoR V and Mlu I restriction sites.
  • PCR polymerase chain reaction
  • the obtained pAVc-PSIL7h (cf.: Figure n°15) is linearized by Pmel, purified on agarose gel and co-transformed with the pAdenoVator ⁇ E1/E3 into BJ5183 competent cells (Q Biogene).
  • the selection for recombinant plasmid-containing cells was on the basis of the antibiotic (kanamycin) resistance marker gene carried on the vector. Plasmid DNA from positives clones were isolated from cultured cells, checked by restriction mapping.
  • the recombinant adenovirus vector is then amplified by transformation of DH5 ⁇ competent cells, plated on LB/kanamycin plates, and midipreparation (Macherey Nagel) of DNA from cell cultures.
  • the sterile Pac I linearized recombinant adenovirus DNA (AdVc-PSIL7h DNA) is used to transfect QBI-293A cells, efficient cells to complement the recombinant adenovirus and produce recombinant viral particules.
  • the human IL-7 encoding cDNA sequence was isolated from the pcDNA-hPSIL-7 by digestion with restriction endonucleases Hind III and BamH I .
  • the resulting "hPSIL-7 cDNA" fragment was purified on agarose gel and inserted, downstream the pCMV promotor, into the pBudCE4.1 vector (Invitrogen) which had been digested with Hind III and BamH I restriction sites.
  • the human BclXL encoding cDNA sequence was amplified by polymerase chain reaction (PCR) ( Mullis et al.; 1987; Methods in Enzymology; 155:, 335-350 ) from human Raji lymphoma cells cDNA (Clontech), using the following oligonucleotides as primers.
  • PCR polymerase chain reaction
  • PCR products were assayed by polyacrylamide or agarose gel electrophoresis in the presence of ethidium bromide and visualization by fluorescence of DNA bands stimulated by UV irradiation.
  • the product band of the size corresponding to the BdXL PCR fragment was isolated and inserted into the plasmid vector pCR II-TOPO (Invitrogen) using the TA-cloning method.
  • the ligation products were transformed into TOP10 competent cells.
  • Plasmid Miniprep Isolation techniques Biorad
  • dideoxy sequencing Sanger et al.; 1977; Proceedings of the National Academy of Sciences of the USA; 74:5463-5467 ) of an asymmetric PCR product DNA using pCR II TOPO universal primers as sequencing primers.
  • pBud-hPSIL-7-BcIXL (cf.: Figure n°17).
  • Example B Fermentation of E. coli producing recombinant IL-7
  • Fermentations for the production of recombinant (human or simian) IL-7 were carried out in 80 liters fermentor (New Brunswick) using an E. coli JM101 host strain transformed with expression plasmid ptac-hIL-7 or ptac-sIL7opt as described in example A1.
  • 1 L inoculum culture was aseptically transferred into fermentor containing 50 L batch medium NRJ18 (pH7).
  • the culture was grown in batch mode (T 37°C, agitation: 100-500 rpm D.O 2 : 30%).
  • the production phase of the fermentation was induced by IPTG (200 mg/L) after D.O 2 reached to 0% until the OD-600 of the culture was about 40.
  • the fermentor content was collected and cooled for at least 30 min in order to reach temperature under 20°C.
  • the culture media was filtered using a 70 ⁇ m filter (PALL R1F-700) to eliminate precipitates and cells were harvested by centrifugation (Beckman J6) at 5000 g for 30 min at 4°C.
  • centrifugation Beckman J6
  • cells will daily be subtracted from the perfusion reactor and induced by tetracycline in a batch reactor.
  • Example C Fermentation of HEK-293 cells producing recombinant hIL-7
  • the best stable positive clone, as in example A2 was adapted to serum-free suspension culture by several media screenings in order to produce a clone optimized for productivity and growth in high cell density culture.
  • Cell culture was performed in a 3 liters bioreactor with a perfusion system. The cells were allowed to grow to a concentration of 10 millions cells/ml. The reactor was operated at a continuous perfusion rate of approximately 3 L/day during 10 days. Roughly 30 L of serum-free culture media containing recombinant protein were generated and used as starting material for the purification of the r-hIL-7.
  • Example D Purification of recombinant IL-7 product Expressed in E. Coli
  • the harvested cells as in example B, were suspended In Tris 20mM / EDTA 10mM buffer (pH8) and centrifuged at 16900 g for 45 min at 4°C. After two successive washing / centrifugation cycles, inclusion bodies fractions were recovered. This inclusion bodies fraction was diluted in order to obtain a protein concentration of 5 to 6 mg/ml and solubilized in solubilization buffer (8 M guanidine hydrochloride - 1 mM EDTA - 1% b-mercaptoethanol - 0.5% DMDAP - 10 mM sodium phosphate - pH8) giving a full reduction, denaturation and solubilization of the protein.
  • solubilization buffer 8 M guanidine hydrochloride - 1 mM EDTA - 1% b-mercaptoethanol - 0.5% DMDAP - 10 mM sodium phosphate - pH8
  • the solution was diluted 1.6 fold in sodium phosphate buffer 6.25 mM and adjusted to 1.5 M ammonium sulfate, pH7.
  • the final concentration of guanidine hydrochloride reached 5M.
  • the solubilized inclusion bodies were pre-filtered and then loaded onto an HIC Butyl 650M (Toso Haas) column equilibrated with the loading buffer (6.25 mM sodium phosphate - 5 M guanidine hydrochloride - 1.7M ammonium sulfate - pH7). After sample application and washing of the column with the same buffer, elution is carried out in one step with 100% of elution buffer (6.25 mM sodium phosphate, 5 M guanidine hydrochloride, pH 7).
  • Renaturated IL-7 was loaded in two times onto either membrane filtration or a G25 Sephadex (Pharmacia) column equilibrated with elution buffer (20 mM sodium phosphate, 0.2 M L-arginine, pH 7) and eluted in order to allow the proper load of r-IL-7 on the subsequent affinity support.
  • the protein peak obtained from the G25 step was loaded onto an Heparin Sepharose Fast Flow (Pharmacia) column equilibrated with a loading buffer (20 mM sodium phosphate, 50 mM sodium chloride, pH 7).
  • elution was carried out in two steps. At first, a fixed ratio of 25 % elution buffer (20 mM sodium phosphate, 1 M sodium chloride, pH 7) / loading buffer was applied over 20 column volumes. Then, refolded IL-7 was eluted in one step with 60 % of elution buffer / loading buffer. The strong selectivity of this step conducted to the elution of the correctly refolded r-IL-7 conformer. In order to eliminate most of the residual impurities including endotoxins, the fraction was adjusted to pH 5 and subjected to a Carboxymethyl Ceramic (BioSepra) column.
  • BioSepra Carboxymethyl Ceramic
  • the CMC column was equilibrated with loading buffer (50 mM sodium acetate, pH 5). After sample application and washing of the column with washing buffer (50 mM sodium acetate, sodium chloride 0.2 M, pH 6), elution was carried out in one step with buffer (50 mM sodium acetate, 0.8 M sodium chloride, pH 6). Finishing steps may also included a G25 Sephadex purification step for desalting followed by a Q Sepharose Fast Flow (QFF Pharmacia) which retained various residual contaminants. R-IL-7 drug substance was recovered pure in the flow through, as showed in figure 9 representing SDS-PAGE analysis: coomassie blue colored and silver stained.
  • Example E Purification of recombinant human IL-7 product Expressed in HEK-293 cells
  • Crude cell culture fluid generated by growth of expression system HEK-293-pcDNA-hPSIL-7 as reported in example C, was treated using a traditional approach or a Streamline ion exchanger mode.
  • crude cell culture fluid was transferred directly from the fermentor to the expanded bed of Streamline ion exchange or heparin, , or Sulfopropyl (SP) or Diethyl Aminoethyl (DEAE), followed by a combination of IEX and HIC. Finishing steps may include filtration and concentration.
  • crude cell culture fluid was clarified using a combination of filtration and concentration [microfiltration (0.45 ⁇ m) ultra/diafiltration] steps to isolate the product.
  • the protein solution obtained was loaded onto an ion exchange combination and Heparin Sepharose [Fast Flow (Pharmacia) column] in various combinations to purify the product.
  • Finishing steps may also included a Hydrophobic Interaction Exchange (HIC) and a filtration/ultrafiltration (UF) or a Carboxymethyl Ceramic (BioSepra) purification step for eliminating residual impurities followed by a G25 Sephadex purification step for desalting followed by a Q Sepharose Fast Flow (QFF Pharmacia) which retained various residual contaminants.
  • HIC Hydrophobic Interaction Exchange
  • UF filtration/ultrafiltration
  • BioSepra Carboxymethyl Ceramic
  • QFF Pharmacia Q Sepharose Fast Flow
  • Example F Product controls and specifications
  • Characterization and proof of structure of this conformer included an amino acid analysis, a peptide mapping after tryptic digest, the sequence of the amino-terminal part, a mass spectrometry (MALDI TOF) control of the molecular weight, a mass spectrometry control of the disulfide bridges, and protein profiles through: SDS PAGE silver stain, Reverse phase HPLC, Cation exchange HPLC, Size exclusion HPLC.
  • MALDI TOF mass spectrometry
  • Example G In vitro biological activity assay of recombinant IL-7:
  • Mammalian (human and simian) IL-7 expressed in E.coli (Example A1), purified and characterized as in example D, were assayed and compared to murin IL-7 (R&D System) for their ability to stimulate the proliferation of the IL-7 dependent cell line designated as pre-B cell line PB1 (DSMZ, Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany) , a cell line derived from bone marrow cells from CBA/C57BL mice.
  • PB1 DSMZ, Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany
  • the PB-1 cell line is absolutely dependent upon exogenous IL-7 for continued growth and viability.
  • the addition of r-IL-7 to theses cultured cell lines stimulates a dose dependent proliferation allowing for a quantitative determination of the levels of r-IL-7 present.
  • the amount of proliferation is measured by "pulsing" each microwell with tritiated thymidine 3H-TdR for 4 hours at 37°C. Dividing pre-B cells will incorporate 3H-Tdr into their DNA.
  • the cells from each well are then harvested onto glass fiber filter discs which trap the DNA.
  • the amount of specifically-bound radioactivity is then measured in a liquid scintillation counter.
  • the number of counts per minute (cpm) for each well is directly proportional to the amount of proliferation by the activated pre-B cells in response to IL-7.
  • Example H Assays for detection of antibodies to recombinant IL-7 in serum:
  • Human recombinant IL-7 is heterologeous for monkey models and may induce neutralizing antibodies against recombinant human IL-7 conformer (as describe above). These antibodies may function as inhibitors in vivo and may contribute to the immunosuppressive therapeutic effects of the drug substance.
  • anti-IL-7-antibodies were investigated in serum and plasma of animals treated with r-hIL-7 or r-sIL-7 pure (according to the present invention) tor r-sIL-7 "impure" as describe in next examples.
  • Methods used for the detection of antibodies in body fluids may included cytokine bioassay, immunometric assay, radioligand assay and various blotting techniques.
  • the plasmatic quantification of anti-r-IL-7 antibodies were achieved using ELISA technique and the serological detection of anti-r-IL-7 antibodies were realized using Western blot technique. These techniques are known to those skilled in the art.
  • Recombinant (human or simian) IL-7 was transferred onto nitrocellulose or PVDF membrane. Then the membrane was incubated with the sera of treated animals (dilution of 1/100 or 1/200) or with anti-IL-7 antibody (Anti-human IL-7 antibody: AB 207 NA, R&D System) for the positive control. After removing the solution, the membrane was washed and incubated with the secondary antibody: Anti-monkey IgG alkaline phosphatase conjugate (SIGMA A1929) or Anti-goat IgG alkaline phosphatase conjugate (SIGMA A4187) for the positive control. Then, the revelation was performed using BCIP/NBT (SIGMA).
  • IL-7 human and simian expressed in E.coli (Example A1), purified and characterized as in example D, and r-sIL-7 "impure" (as defined in example G) were tested for in vivo biological activity in normal primates.
  • This primate model was the only possible model to test the long term activity of the drug substance and the immunogenicity of the same drug substance contaminated with molecular variants and/or drug product related impurities.
  • Peripheral blood specimens were collected from animals under ketamine restraint before and after IL-7 treatment (on days 0 (pretreatment), 7, 14, 21, 28, 35 and 42).
  • CD4 cellularity was examined on a FACScan cell analysis system (Becton Dickinson). Serum samples were examined for the presence of antibodies by western blot analysis and confirmed by ELISA analysis, as describe in example H.
  • Blood CD4 T cell count is indicated in figure 11 : represented as a median of blood CD4 T cell count for each group. All animals survived the protocol and tolerated IL-7 administration without adverse reactions to IL-7 therapy.
  • peripheral blood CD4 T cell count increased 1.4 to 2.1 X from day 14 until day 35 and remain above pretreatment values at day 42 in group 3 (treated with r-sIL-7) compared to placebo group and CD4 cellularity began to increase and then decreased from week 2 until week 6 for groups 2 (treated with r-hIL-7) and 4 (treated with rsIL-7 "impure” preparation).
  • Anti-IL-7 antibodies were detected in the sera of these two groups of monkeys (2 and 4), during the second week after receiving r-hIL-7 and r-sIL-7 "impure” respectively, by subcutaneous injection (cf. figure 12 ). In these normal monkeys, the repeated injection of heterologous or impure IL-7 drug substance did trigger the appearance of anti-IL-7 antibodies and simultaneously this produce a strong decrease of the IL-7 lymphopoietic effect.
  • simian IL-7 expressed in E.coli (Example A1), purified and characterized as in example D, and r-sIL-7 "impure" (as defined in example G) were tested for mid or long term in vivo biological activity in immunodepressed primates. Young rhesus monkeys were studied in three groups (1 to 3), each group contained 3 animals. All monkeys underwent total body irradiation (TBI) (6.1 Gy). Monkeys received following regimens:
  • Example K In vivo activity of recombinant human IL-7 on pharmacodynamic parameters and total lymphocyte count in normal cynomolgus monkeys
  • Recombinant human IL-7 (r-hlL-7) expressed in E. coli (Example A1), purified and characterized as in example D, was tested for in vivo biological activity on pharmacodynamic parameters and total lymphocyte count in normal cynomolgus monkeys.
  • Four normal female Cynomolgus monkeys received subcutaneous bolus single injection of soluble r-hlL-7 at the dose level of 100 ⁇ g/kg. All animals were studied over 96 hours. Laboratory investigations (hematology and immune cell phenotyping) were performed before r-hlL-7 single injection and 6, 24, 48, 72 and 96 hours post-injection.
  • lymphocyte subsets were examined by Flow Cytometry using highly specific cell surface antibody markers including: CD3, CD4, CD8, CD20, CD127, Ki67 and Bcl2 in various combinations. Observed variations in lymphocyte count and specific markers were reported in the next table and in histogram referred as figure ...: Lympho 10 ⁇ 6/ml CD20% CD3% CD4% CD8% K67% CD127% BCL-2% before 8,929 14,75 36,80 10,94 38,23 1,93 40,86 21,61 day 1 (+6hs) 4,823 24,90 21,48 6,36 23,83 2,39 6,95 11,03 day 2 4,851 12,80 27,63 8,45 26,82 3,41 4,55 52,15 day 3 4,880 8,55 27,88 5,75 32,40 3,81 8,10 49,30 day 4 6,441 13,45 58,25 19,85 48,83 7,71 23,43 65,63 day 5 8,003 9,33 45,33 11,23 43,50 5,21 42,53 30,23

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pulmonology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biotechnology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • AIDS & HIV (AREA)
  • Transplantation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Claims (40)

  1. Eine pharmazeutische Zusammensetzung, umfassend IL-7 und eine oder mehrere pharmazeutisch kompatible oder verträgliche Träger, Hilfsstoffe oder Verdünnungsmittel, wobei mindestens 98 Gewichts-% der Gesamtmenge an IL-7 in der Zusammensetzung aus einem IL-7 Konformer besteht, das die folgenden drei Disulfidbrücken umfasst: Cys: 1-4 (Cys2-Cys92); 2-5 (Cys34-Cys129) und 3-6 (Cys47-Cys141).
  2. Die Zusammensetzung nach Anspruch 1, wobei das IL-7 Konformer ein rekombinates humanes IL-7 Konformer ist.
  3. Die Zusammensetzung nach Anspruch 2, wobei das IL-7 Konformer die Aminosäuresequenz der SEQ ID NO: 2 oder 4 umfasst.
  4. Die Zusammensetzung nach Anspruch 1, wobei das IL-7 Konformer ein rekombinates IL-7 Konformer vom Affe ist.
  5. Die Zusammensetzung nach Anspruch 4, wobei das IL-7 Konformer die Aminosäuresequenz der SEQ ID NO: 12 umfasst.
  6. Die Zusammensetzung nach einem der Ansprüche 1 bis 5, wobei das IL-7 Konformer nicht glykosyliert ist.
  7. Die Zusammensetzung nach einem der Ansprüche 1 bis 5, wobei das IL-7 Konformer glykosyliert ist.
  8. Die Zusammensetzung nach einem der Ansprüche 1 bis 7, wobei das IL-7 Konformer mit dem Hepatozytenwachstumsfaktor als Heterodimer assoziiert ist.
  9. Die Zusammensetzung nach einem der Ansprüche 1 bis 7, wobei das IL-7 Konformer funktionell an einen Fc-Teil einer IgG schweren Kette durch eine Peptidgelenkregion angehängt ist, wobei das IgG ein humanes IgG1 oder IgG4 ist.
  10. Die Zusammensetzung nach einem der Ansprüche 1 bis 7, wobei das IL-7 Konformer funktionell an ein humanes Serum Albumin (HSA) oder einen Teil von HSA als Fusionsprotein angehängt ist.
  11. Die Zusammensetzung nach einem der Ansprüche 1 bis 10, die frei von anderen IL-7 Konformeren ist.
  12. Die Zusammensetzung nach einem der Ansprüche 1 bis 11, wobei die Gesamtmenge pro Gewicht des IL-7 Konformers mindestens 99,5 Gewichts-% der Gesamtmenge an IL-7 in der Zusammensetzung ist.
  13. Die Zusammensetzung nach einem der Ansprüche 1 bis 12, die einen pharmazeutisch kompatiblen Träger umfasst, der ausgewählt ist aus Sucrose, Trehalose und einer Aminosäure.
  14. Die Zusammensetzung nach einem der Ansprüche 1 bis 13, die einen pharmazeutisch kompatiblen Träger umfasst, der in einem geeigneten Puffer enthalten ist, um eine isotonische Lösung zu bilden.
  15. Die Zusammensetzung nach Anspruch 14, wobei der geeignete Puffer einen pH-Bereich hat, der zwischen 5 bis 7,5, vorzugsweise 6 bis 7, noch bevorzugter 6,5 umfasst ist.
  16. Die Zusammensetzung nach Anspruch 15, wobei der geeignete Puffer ein organisches Salz ist, das ausgewählt ist aus einem Natriumzitratpuffer und einem Ammoniumazetatpuffer.
  17. Die Zusammensetzung nach einem der Ansprüche 1 bis 16, wobei die Zusammensetzung in lyophilisierter Form vorliegt.
  18. Die Zusammensetzung nach einem der Ansprüche 1 bis 17, wobei die Zusammensetzung ein Protein (vorzugsweise humanes Serumalbumin) und/oder eine oberflächenaktive Substanz (vorzugsweise Tween 80) umfasst.
  19. Die Zusammensetzung nach einem der Ansprüche 11 bis 18, wobei die Zusammensetzung weiterhin ein immunstimulierendes Agens umfasst, das ausgewählt ist aus einem hämatopoietischen Zellwachstumsfaktor, einem Zytokin, einem Antigen und einem Adjuvans, oder einer Kombination davon, für eine kombinierte, getrennte oder sequenzielle Verwendung.
  20. Die Zusammensetzung nach Anspruch 19, wobei der hämatopoietische Zellwachstumsfaktor ausgewählt ist aus dem Stammzellfaktor (SCF), insbesondere der löslichen Form des SCF, G-CSF, GM-CSF, Flt-3 Ligand, IL-15 und IL-2.
  21. Die Zusammensetzung nach Anspruch 19, wobei das Zytokin ausgewählt ist aus - Interferon, IL-2, IL-12, RANTES, B7-1, MIP-2 und MIP-1α.
  22. Die Zusammensetzung nach einem der Ansprüche 19 bis 21, wobei das Antigen ausgewählt ist aus einem synthetischen oder natürlichen Peptid, einem rekombinanten Protein, einem abgetöteten, inaktivierten oder abgeschwächten pathogenen Produkt, einem Lipid, einem Teil davon und einer Kombination davon.
  23. Die Zusammensetzung nach Anspruch 22, wobei das Antigen ausgewählt ist aus Antigenen, die vom HIV, Varicella-Zoster-Virus, Influenzavirus, Epstein-Barr-Virus, Typ 1 oder 2 Herpes-Simplex-Virus, humanen Zytomegalovirus, Denge-Virus, Hepatitis A, B, C oder E-Virus, respiratorischen Synzytialvirus, humanen Papillomavirus, Mycobacterium Tuberculosis, Toxoplasma und Chlamydia abstammen.
  24. Die Zusammensetzung nach einem der Ansprüche 19 bis 23, wobei das Adjuvans ausgewählt ist aus irgendeiner Substanz, Gemisch, Lösung oder Zusammensetzung, die die Immunogenität eines Antigens beschleunigt oder erhöht und fähig ist, eine Th-1Typ Immunantwort zu induzieren, wie CpG, QS21, ISCOM und Monophosphoryl-Lipid A.
  25. Pharmazeutische Zusammensetzung nach einem der Ansprüche 1 bis 24, zur Verabreichung an einen menschlichen Patienten zur prophylaktischen oder therapeutischen Stimulation der Entwicklung und Proliferation von B- oder T-Lymphozyten, oder zur Verstärkung der globalen oder spezifischen Immunrekonstitution, oder zur Verstärkung der humoralen oder zellulären Immunantwort.
  26. Eine pharmazeutische Zusammensetzung nach einem der Ansprüche 1 bis 24, um opportunistische Infektionen in immundeffizienten Patienten zu verhindern oder zu reduzieren.
  27. Eine pharmazeutische Zusammensetzung nach einem der Ansprüche 1 bis 24, um die Stimulation der Lymphopoese auszudehnen und/oder spezifische Immunantworten auszulösen und/oder das Repertoire einer spezifischen Immunantwort in menschlichen Patienten zu erweitern.
  28. Eine pharmazeutische Zusammensetzung nach den Ansprüchen 25, 26 oder 27, zur Verwendung in menschlichen Patienten, die immundeffiziente Patienten, Krebspatienten sind, Patienten, die sich Transplantationen unterziehen, Patienten, die mit einem Virus oder einem Parasit infiziert sind, ältere Patienten oder jeder Patient mit einer niedrigen CD4 Zahl.
  29. Die Zusammensetzung nach einem der Ansprüche 1 bis 28, zur Verwendung bei einer effektiven Menge an IL-7, die zwischen ungefähr 3 bis 300µg/kg/Tag, vorzugsweise zwischen 10 bis 100 µg/kg/Tag, umfasst ist, und insbesondere von einmal täglich bis zweimal oder dreimal pro Woche bis hinunter auf einmal pro Woche verabreicht wird.
  30. Ein Verfahren zur Herstellung einer pharmazeutischen Zusammensetzung, wie in einem der Ansprüche 1 bis 29 definiert, wobei das Verfahren umfasst:
    a) Bereitstellen einer Probe, die IL-7 Polypeptide umfasst.
    b) Reinigen eines IL-7 Konformers, das die folgenden drei Disulfidbrücken umfasst: Cys: 1-4 (Cys2-Cys92); 2-5 (Cys34-Cys129) und 3-6 (Cys47-Cys141) um eine IL-7 Arzneimittelsubstanz zu produzieren, und
    c) wahlweise messen oder quantifizieren des bestimmten IL-7 Konformers in der Arzneimittelsubstanz.
  31. Das Verfahren nach Anspruch 30, wobei die Probe von rekombinanten prokaryotischen und eukaryotischen Wirtszellen erhalten wird, die IL-7 Polypetide produzieren.
  32. Das Verfahren nach Anspruch 31, wobei die Probe eine Kultur prokaryotischer Wirtszellen ist (oder von einer abstammt), die ein IL-7 Polypeptid kodieren und weiterhin, wobei das Verfahren weiterhin vor Schritt b) umfasst:
    i) Behandeln der Probe, um eine vollständige Denaturierung der IL-7 Polypeptide zu verursachen.
    ii) wahlweise Reinigen der denaturierten Polypeptide, die in Schritt i) erhalten wurden und
    iii) Rückfalten der Polypeptide.
  33. Das Verfahren nach Anspruch 32, wobei Schritt i) das Lösen von Einschlusskörperchen in einem Denaturierungspuffer umfasst.
  34. Das Verfahren nach Anspruch 32 oder 33, wobei Schritt ii) mittels hydrophober Chromatographie, Ionenaustausch- oder Umkehrphasen-Chromatographie durchgeführt wird.
  35. Das Verfahren nach Anspruch 33, wobei die hydrophobe Chromatographie unter Verwendung von HIC Butyl angewendet wird.
  36. Das Verfahren nach einem der Ansprüche 32 bis 35, wobei Schritt ii) bei einem pH durchgeführt wird, der zwischen 6 und 9, vorzugsweise zwischen 7 und 8,5 einschließlich umfasst ist.
  37. Das Verfahren nach einem der Ansprüche 32 bis 36, wobei der Reinigungsschritt b das Durchführen einer Affinitätschromatographie umfasst.
  38. Das Verfahren nach Anspruch 37, wobei die Affinitätschromatographie auf einer Säule aus Sulfatpolysacchariden durchgeführt wird.
  39. Das Verfahren nach Anspruch 38, wobei das Sulfatpolysaccharid Dextransulfat oder Heparin ist.
  40. Das Verfahren nach einem der Ansprüche 30 bis 39, wobei das IL-7 Konformer in der Arzneimittelsubstanz charakterisiert wird durch Massenspektrometrie, Infrarotspektroskopie, NMR, durch Bestimmen des zirkularen Dichronismus, durch Messen der Affinität gegenüber einem spezifischen monoklonalen Antikörper, der gegen das IL-7 Konformer gerichtet ist, oder durch Heparin-Affinitätschromatographie, und gemessen oder quantifiziert wird durch ELISA, Bioassay oder die Affinität des IL-7 Konformers für den IL-7 Rezeptor und jedes Verfahren der Proteinquantifikation, das auf das isolierte Konformer angewendet wird.
EP03792262A 2002-08-08 2003-08-06 Il-7 als wirkstoff, il-7-haltige zusammensetzung, herstellungsverfahren und verwendungen Expired - Lifetime EP1527179B1 (de)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP03792262A EP1527179B1 (de) 2002-08-08 2003-08-06 Il-7 als wirkstoff, il-7-haltige zusammensetzung, herstellungsverfahren und verwendungen
SI200331907T SI1527179T1 (sl) 2002-08-08 2003-08-06 Il-7 zdravilna substanca il-7 vsebujoäś sestavek, njena priprava in uporaba
CY20101101165T CY1110994T1 (el) 2002-08-08 2010-12-17 Il-7 φαρμακευτικη ουσια, συνθεση που περιεχει il-7 παρασκευη και χρησεις αυτης

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP02291996A EP1391513A1 (de) 2002-08-08 2002-08-08 IL-7 als Wirkstoff, IL-7-haltige Zusammensetzung, Herstellungsverfahren und Verwendungen
EP02291996 2002-08-08
US47588103P 2003-06-05 2003-06-05
US475881P 2003-06-05
PCT/EP2003/008701 WO2004018681A2 (en) 2002-08-08 2003-08-06 Il-7 drug substance, il- 7 comprising composition, preparation and uses thereof
EP03792262A EP1527179B1 (de) 2002-08-08 2003-08-06 Il-7 als wirkstoff, il-7-haltige zusammensetzung, herstellungsverfahren und verwendungen

Publications (2)

Publication Number Publication Date
EP1527179A2 EP1527179A2 (de) 2005-05-04
EP1527179B1 true EP1527179B1 (de) 2010-09-22

Family

ID=30775891

Family Applications (2)

Application Number Title Priority Date Filing Date
EP02291996A Withdrawn EP1391513A1 (de) 2002-08-08 2002-08-08 IL-7 als Wirkstoff, IL-7-haltige Zusammensetzung, Herstellungsverfahren und Verwendungen
EP03792262A Expired - Lifetime EP1527179B1 (de) 2002-08-08 2003-08-06 Il-7 als wirkstoff, il-7-haltige zusammensetzung, herstellungsverfahren und verwendungen

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP02291996A Withdrawn EP1391513A1 (de) 2002-08-08 2002-08-08 IL-7 als Wirkstoff, IL-7-haltige Zusammensetzung, Herstellungsverfahren und Verwendungen

Country Status (18)

Country Link
US (1) US7585947B2 (de)
EP (2) EP1391513A1 (de)
JP (3) JP2005534339A (de)
AT (1) ATE482273T1 (de)
AU (1) AU2003250216B2 (de)
CA (1) CA2494974C (de)
CY (1) CY1110994T1 (de)
DE (1) DE60334301D1 (de)
DK (1) DK1527179T3 (de)
ES (1) ES2353006T3 (de)
HK (1) HK1075465A1 (de)
IL (1) IL166543A (de)
NO (1) NO332305B1 (de)
PL (1) PL213710B1 (de)
PT (1) PT1527179E (de)
SI (1) SI1527179T1 (de)
WO (1) WO2004018681A2 (de)
ZA (1) ZA200501914B (de)

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2367891T3 (es) 2000-09-29 2011-11-10 Schering Corporation Interleucina-10 pegilada.
CA2487673C (en) * 2003-12-02 2010-11-02 F. Hoffmann-La Roche Ag Improved method for the recombinant production and purification of protein kinases
RU2369616C2 (ru) * 2003-12-30 2009-10-10 Мерк Патент Гмбх Слитые белки il-7
EP1746161A1 (de) * 2005-07-20 2007-01-24 Cytheris Glykosylierte IL-7, Verfahren zur Herstellung und Verwendung davon
CA2619989C (en) * 2005-08-23 2014-06-17 National Research Council Of Canada Regulation of heterologous recombinant protein expression in methylotrophic and methanotrophic bacteria
EP3431585A1 (de) 2006-05-31 2019-01-23 The Regents of The University of California Cd127-expression, die invers mit foxp3 und der suppressiven funktion von cd4+-tregs korreliert
BRPI0719446A2 (pt) 2006-09-28 2013-12-10 Schering Corp Uso de il-10 peguilada para tratar câncer
EP2649094B1 (de) 2010-12-10 2016-04-27 University of Central Florida Research Foundation, Inc. Verfahren und zusammensetzungen mit il-7 rezeptor-liganden
KR20140063657A (ko) 2011-08-03 2014-05-27 사이세리스 Hcv 면역요법
CN110227152A (zh) * 2012-04-23 2019-09-13 巴拉特生物技术国际有限公司 轮状病毒疫苗组合物及其制备方法
RU2562169C2 (ru) * 2012-10-29 2015-09-10 Федеральное государственное унитарное предприятие "Государственный научно-исследовательский институт особо чистых биопрепаратов" Федерального медико-биологического агентства Штамм культивируемых клеток cho-il7/13 - продуцент интерлейкина-7 человека
JP2016519108A (ja) 2013-04-18 2016-06-30 アルモ・バイオサイエンシーズ・インコーポレイテッド インターロイキン−10を疾病及び疾患の治療に用いる方法
WO2014204816A2 (en) 2013-06-17 2014-12-24 Armo Biosciences, Inc. Method for assessing protein identity and stability
KR20160079114A (ko) 2013-11-11 2016-07-05 아르모 바이오사이언시스 인코포레이티드 질환 및 장애를 치료하기 위한 인터류킨-10을 사용하는 방법
EP3119412A1 (de) 2014-03-21 2017-01-25 Boreal Invest Endgültige nanofiltration von gelösten proteinzusammensetzungen zur entfernung von immunogenen aggregaten
WO2015187295A2 (en) 2014-06-02 2015-12-10 Armo Biosciences, Inc. Methods of lowering serum cholesterol
WO2016064817A1 (en) 2014-10-22 2016-04-28 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
US10618970B2 (en) 2015-02-03 2020-04-14 Armo Biosciences, Inc. Method of treating cancer with IL-10 and antibodies that induce ADCC
WO2016145388A1 (en) * 2015-03-11 2016-09-15 Nektar Therapeutics Conjugates of an il-7 moiety and an polymer
AU2016268403A1 (en) 2015-05-28 2017-12-07 Armo Biosciences, Inc. Pegylated interleukin-10 for use in treating cancer
EP3307766A4 (de) 2015-06-11 2019-06-12 Genexine, Inc. Modifiziertes interleukin-7-protein und verwendungen davon
KR102386735B1 (ko) 2015-11-06 2022-04-14 주식회사 제넥신 변형된 인터루킨-7 융합 단백질의 제형
RU2615447C1 (ru) * 2015-11-13 2017-04-04 Федеральное государственное унитарное предприятие "Государственный научно-исследовательский институт особо чистых биопрепаратов" Федерального медико-биологического агентства Синтетическая ДНК, кодирующая интерлейкин-7 человека, содержащий ее экспрессионный вектор (варианты), штамм-продуцент интерлейкина-7 человека и способ получения интерлейкина-7 человека
WO2017095191A1 (ko) * 2015-12-04 2017-06-08 주식회사 제넥신 면역글로불린 fc가 융합된 인터루킨-7 융합 단백질을 포함하는 사람 파필로마바이러스 유래 질환의 예방 또는 치료용 약학적 조성물
US11357827B2 (en) 2015-12-04 2022-06-14 Genexine, Inc. Method for preventing or treating influenza virus infection using pharmaceutical composition comprising immunoglobulin Fc-fused interleukin-7 fusion protein
WO2018156649A1 (en) * 2017-02-22 2018-08-30 Flagship Pioneering, Inc. Compositions of t cell modulator (tcm) molecules and uses thereof
CA3119341A1 (en) 2018-11-16 2020-05-22 Neoimmunetech, Inc. Method of treating a tumor with a combination of il-7 protein and an immune checkpoint inhibitor
JP2022514702A (ja) 2018-12-21 2022-02-14 オーエスイー・イミュノセラピューティクス 二機能性抗pd-1/il-7分子
AU2020383176B2 (en) * 2019-11-15 2023-10-19 Genexine, Inc. Fusion protein including modified interleukin-7 and TGF beta receptor II and use thereof
AU2021207586A1 (en) 2020-01-13 2022-07-21 Neoimmunetech, Inc. Method of treating a tumor with a combination of IL-7 protein and a bispecific antibody
JP2023512657A (ja) 2020-02-05 2023-03-28 ワシントン・ユニバーシティ Il-7タンパク質とcar保有免疫細胞の組み合わせで固形腫瘍を治療する方法
WO2022093718A1 (en) 2020-10-26 2022-05-05 Neoimmunetech, Inc. Methods of inducing stem cell mobilization
WO2022094475A1 (en) 2020-11-02 2022-05-05 Neoimmunetech, Inc. Use of interleukin-7 for the treatment of coronavirus
KR20230104175A (ko) 2020-11-05 2023-07-07 네오이뮨텍, 인코퍼레이티드 Il-7 단백질과 뉴클레오타이드 백신의 조합물을 사용한 종양의 치료 방법
WO2022117569A1 (en) 2020-12-02 2022-06-09 Oncurious Nv A ccr8 antagonist antibody in combination with a lymphotoxin beta receptor agonist antibody in therapy against cancer
WO2023130081A1 (en) 2021-12-30 2023-07-06 Neoimmunetech, Inc. Method of treating a tumor with a combination of il-7 protein and vegf antagonist
WO2023133595A2 (en) 2022-01-10 2023-07-13 Sana Biotechnology, Inc. Methods of ex vivo dosing and administration of lipid particles or viral vectors and related systems and uses
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2024102722A1 (en) 2022-11-07 2024-05-16 Neoimmunetech, Inc. Methods of treating a tumor with an unmethylated mgmt promoter
CN117050178B (zh) * 2023-10-13 2024-01-12 北京百普赛斯生物科技股份有限公司 特异性检测il-7的抗体及应用

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA887773B (en) * 1987-10-26 1989-07-26 Immunex Corp Interleukin-7
US4965195A (en) * 1987-10-26 1990-10-23 Immunex Corp. Interleukin-7
US5328988A (en) * 1987-10-26 1994-07-12 Immunex Corporation Interleukin-7
US5714585A (en) * 1987-10-26 1998-02-03 Sterling Winthrop, Inc. Antibodies that are immunoreactive with interleukin-7
US5728680A (en) * 1987-12-30 1998-03-17 Cytoven J.V. Methods for normalizing numbers of lymphocytes
US5459058A (en) * 1991-03-28 1995-10-17 Benjamin Rich Cell culture system
US5223408A (en) * 1991-07-11 1993-06-29 Genentech, Inc. Method for making variant secreted proteins with altered properties
WO1994022473A1 (en) * 1993-04-01 1994-10-13 University Of Washington Use of interleukin 7 to improve vaccine potency
US5696234A (en) * 1994-08-01 1997-12-09 Schering Corporation Muteins of mammalian cytokine interleukin-13
EP1012184B1 (de) * 1997-07-14 2007-10-10 Bolder Biotechnology, Inc. Derivate des wachstumshormons und verwandte proteine
AU6246199A (en) * 1998-09-21 2000-04-10 Schering Corporation Human interleukin-b50, therapeutic uses
CA2404479A1 (en) * 2000-03-30 2001-10-11 University Of Connecticut Hybrid cytokine of il-7 and .beta.-chain of hepatocyte growth factor
RU2369616C2 (ru) * 2003-12-30 2009-10-10 Мерк Патент Гмбх Слитые белки il-7
EP1746161A1 (de) * 2005-07-20 2007-01-24 Cytheris Glykosylierte IL-7, Verfahren zur Herstellung und Verwendung davon

Also Published As

Publication number Publication date
ATE482273T1 (de) 2010-10-15
ZA200501914B (en) 2005-11-30
CA2494974A1 (en) 2004-03-04
NO332305B1 (no) 2012-08-20
IL166543A0 (en) 2006-01-15
JP2005534339A (ja) 2005-11-17
JP2010115203A (ja) 2010-05-27
US20050249701A1 (en) 2005-11-10
HK1075465A1 (en) 2005-12-16
US7585947B2 (en) 2009-09-08
AU2003250216A1 (en) 2004-03-11
AU2003250216B2 (en) 2010-08-05
WO2004018681A2 (en) 2004-03-04
NO20050355L (no) 2005-05-06
ES2353006T3 (es) 2011-02-24
EP1527179A2 (de) 2005-05-04
WO2004018681A3 (en) 2004-04-01
CA2494974C (en) 2014-07-29
JP2014147396A (ja) 2014-08-21
DK1527179T3 (da) 2011-01-03
PL373606A1 (en) 2005-09-05
EP1391513A1 (de) 2004-02-25
PL213710B1 (pl) 2013-04-30
PT1527179E (pt) 2010-12-07
DE60334301D1 (de) 2010-11-04
SI1527179T1 (sl) 2011-04-29
IL166543A (en) 2012-02-29
JP5980467B2 (ja) 2016-08-31
CY1110994T1 (el) 2015-06-11

Similar Documents

Publication Publication Date Title
EP1527179B1 (de) Il-7 als wirkstoff, il-7-haltige zusammensetzung, herstellungsverfahren und verwendungen
US8153114B2 (en) Hyperglycosylated mammalian IL-7 IgG conjugate composition
AU2004309050B2 (en) IL-7 fusion proteins

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20050131

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1075465

Country of ref document: HK

17Q First examination report despatched

Effective date: 20080214

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CYTHERIS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: PATENTANWAELTE SCHAAD, BALASS, MENZL & PARTNER AG

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 60334301

Country of ref document: DE

Date of ref document: 20101104

Kind code of ref document: P

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20101129

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Effective date: 20110214

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 8652

Country of ref document: SK

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E009794

Country of ref document: HU

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1075465

Country of ref document: HK

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20110623

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 60334301

Country of ref document: DE

Effective date: 20110623

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CY

Payment date: 20150803

Year of fee payment: 13

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20100403008

Country of ref document: GR

Effective date: 20110119

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160831

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 14

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160807

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MC

Payment date: 20170301

Year of fee payment: 14

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160806

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160831

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BG

Payment date: 20170206

Year of fee payment: 14

PGRI Patent reinstated in contracting state [announced from national office to epo]

Ref country code: BE

Effective date: 20170301

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 15

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170831

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 16

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180306

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20220830

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20220729

Year of fee payment: 20

Ref country code: SK

Payment date: 20220801

Year of fee payment: 20

Ref country code: SE

Payment date: 20220831

Year of fee payment: 20

Ref country code: RO

Payment date: 20220801

Year of fee payment: 20

Ref country code: PT

Payment date: 20220728

Year of fee payment: 20

Ref country code: LU

Payment date: 20220830

Year of fee payment: 20

Ref country code: IT

Payment date: 20220831

Year of fee payment: 20

Ref country code: IE

Payment date: 20220829

Year of fee payment: 20

Ref country code: GB

Payment date: 20220830

Year of fee payment: 20

Ref country code: FI

Payment date: 20220829

Year of fee payment: 20

Ref country code: ES

Payment date: 20220908

Year of fee payment: 20

Ref country code: EE

Payment date: 20220824

Year of fee payment: 20

Ref country code: DK

Payment date: 20220831

Year of fee payment: 20

Ref country code: DE

Payment date: 20220826

Year of fee payment: 20

Ref country code: CZ

Payment date: 20220805

Year of fee payment: 20

Ref country code: AT

Payment date: 20220829

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SI

Payment date: 20220729

Year of fee payment: 20

Ref country code: HU

Payment date: 20220801

Year of fee payment: 20

Ref country code: GR

Payment date: 20220826

Year of fee payment: 20

Ref country code: FR

Payment date: 20220805

Year of fee payment: 20

Ref country code: BE

Payment date: 20220830

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20220907

Year of fee payment: 20

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230524

REG Reference to a national code

Ref country code: DE

Ref legal event code: R071

Ref document number: 60334301

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EUP

Expiry date: 20230806

REG Reference to a national code

Ref country code: NL

Ref legal event code: MK

Effective date: 20230805

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: SK

Ref legal event code: MK4A

Ref document number: E 8652

Country of ref document: SK

Expiry date: 20230806

REG Reference to a national code

Ref country code: BE

Ref legal event code: MK

Effective date: 20230806

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20230828

REG Reference to a national code

Ref country code: GB

Ref legal event code: PE20

Expiry date: 20230805

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK07

Ref document number: 482273

Country of ref document: AT

Kind code of ref document: T

Effective date: 20230806

REG Reference to a national code

Ref country code: IE

Ref legal event code: MK9A

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20230817

Ref country code: IE

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20230806

Ref country code: GB

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20230805

Ref country code: ES

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20230807

Ref country code: CZ

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20230806

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20230806

Ref country code: SI

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20230807